Language selection

Search

Patent 2688036 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent: (11) CA 2688036
(54) English Title: FORMULATIONS FOR THE ORAL ADMINISTRATION OF THERAPEUTIC AGENTS AND RELATED METHODS
(54) French Title: FORMULATIONS POUR L'ADMINISTRATION PAR VOIE ORALE D'AGENTS THERAPEUTIQUES, ET PROCEDES ASSOCIES
Status: Granted
Bibliographic Data
(51) International Patent Classification (IPC):
  • A61K 47/24 (2006.01)
  • A61K 9/107 (2006.01)
  • A61K 31/7048 (2006.01)
  • A61K 45/06 (2006.01)
  • A61K 45/08 (2006.01)
  • A61K 47/14 (2017.01)
  • A61P 31/10 (2006.01)
  • A61P 33/02 (2006.01)
(72) Inventors :
  • WASAN, ELLEN K. (Canada)
  • WASAN, KISHOR M. (Canada)
(73) Owners :
  • THE UNIVERSITY OF BRITISH COLUMBIA (Canada)
(71) Applicants :
  • THE UNIVERSITY OF BRITISH COLUMBIA (Canada)
(74) Agent: BERESKIN & PARR LLP/S.E.N.C.R.L.,S.R.L.
(74) Associate agent:
(45) Issued: 2016-08-16
(86) PCT Filing Date: 2008-05-23
(87) Open to Public Inspection: 2008-12-04
Examination requested: 2013-05-22
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/CA2008/000975
(87) International Publication Number: WO2008/144888
(85) National Entry: 2009-11-24

(30) Application Priority Data:
Application No. Country/Territory Date
60/940,307 United States of America 2007-05-25
60/976,708 United States of America 2007-10-01
61/041,478 United States of America 2008-04-01

Abstracts

English Abstract

The present application relates to an oral formulation of amphotericin B and other therapeutic agents, which formulation comprises one or more fatty acid glycerol esters and one or more PEG modified phospholipids or fatty acid esters. The formulation provides enhanced bioavailability and/or increased stability of the therapeutic agent at the low pH found in gastric fluid.


French Abstract

La présente invention concerne une formulation orale d'amphotéricine B et d'autres agents thérapeutiques qui comprend au moins un ester de glycérol d'acide gras et au moins un phospholipide modifié par PEG ou des esters d'acide gras. La formulation engendre une biodisponibilité améliorée et/ou une stabilité accrue de l'agent thérapeutique à un niveau du pH faible propre au liquide gastrique.

Claims

Note: Claims are shown in the official language in which they were submitted.


CLAIMS
1. Amphotericin B formulation, comprising,
(a) amphotericin B;
(b) one or more fatty acid glycerol esters;
(c) one or more polyethylene oxide-containing fatty acid esters; and
optionally
(d) glycerol in an amount less than 10% by weight,
wherein the ratio of fatty acid glycerol esters to polyethylene
oxide-containing fatty acid esters is from about 20:80 to about 80:20 v/v.
2. Formulation of claim 1 wherein the amphotericin B is in an amount from
about 0.5 to about 10 mg/mL of the formulation.
3. Formulation of claim 1 or 2, wherein the fatty acid glycerol esters
comprise from about 32 to about 52% by weight fatty acid monoglycerides, from
about
30 to about 50% by weight fatty acid diglycerides, from about 5 to about 20%
by weight
fatty acid triglycerides, and greater than 60% by weight oleic acid mono-, di-
, and
triglycerides.
4. Formulation of any one of claims 1 to 3, wherein the polyethylene
oxide-containing fatty acid esters comprise a polyethylene oxide ester of a C8-
C22
saturated fatty acid.
5. Formulation of claim 4 wherein the polyethylene oxide-containing fatty
acid esters comprise a polyethylene oxide ester of a C12-C18 saturated fatty
acid.
6. Formulation of claim 4 or 5 wherein the polyethylene oxide-containing
fatty acid esters are selected from the group consisting of lauric acid
esters, palmitic acid
esters, stearic acid esters, and mixtures thereof.
-47-

7. Formulation of any one of claims 4 to 6 wherein the polyethylene
oxide-containing fatty acid esters comprise a polyethylene oxide having an
average
molecular weight of from about 750 to about 2000.
8. Formulation of any one of claims 1 to 7, wherein the formulation is a
self-emulsifying drug delivery system.
9. Use of a formulation of any one of claims 1 to 8 for administering
amphotericin B to a subject in need thereof.
10. Use of a therapeutically effective amount of an amphotericin B
formulation of any one of claims 1 to 8 for treating an infectious disease
treatable by the
administration of amphotericin B in a subject in need thereof.
11. Use of claim 9 or 10, wherein the formulation is for oral or topical
use.
12. Use of claim 10, wherein the infectious disease is a fungal infection
selected from the group consisting of aspergillosis, blastomycosis,
candidiasis,
coccidioidomycosis, crytococcosis, histoplasmosis,
mucormycosis,
paracoccidioidomycosis, sporotrichosis, visceral leishmaniasis, cutaneous
leishmaniasis,
Chagas disease, and Febrile neutropenia.
-48-

Description

Note: Descriptions are shown in the official language in which they were submitted.


CA 02688036 2015-05-20
WO 2008/144888 PCT/CA2008/000975
FORMULATIONS FOR THE ORAL ADMINISTRATION
OF THERAPEUTIC AGENTS AND RELATED METHODS
BACKGROUND OF THE INVENTION
Each year in the Indian subcontinent alone, over 500,000 individuals play host
to
Leishmania donovani, an insidious parasite that invades macrophages, rapidly
infiltrates
the vital organs and ultimately leads to severe infection of the visceral
reticuloendothelial
system. Visceral leishmaniasis, also known as Kala-azar, is most prevalent in
the weak
and the young within a population. Left untreated, almost all infected
individuals will
die. Visceral leishmaniasis affects over 200 million people from 62 countries.
The
therapeutic arsenal against Leishmania is limited to a small number of
parenterally
administered agents, with daily injections of pentavalent antimony compound.
Although
more expensive than the antimonials, amphotericin B (AmpB) has a 97% cure rate
and no
reported resistance. However, drug therapy involves IV administration over 30-
40 days
and is associated with infusion-related side-effects (fever, chills, bone
pain,
thrombophlebitis). The dose-limiting toxicity, which may even affect the
ability to
achieve a cure, is renal impairment. In addition, due to the prohibitive cost
and difficult
route of drug administration, amphotericin B is failing to reach many
patients.
In developed nations, disseminated fungal infections such as candidiasis,
histoplasmosis, coccidiosis, and aspergillosis are on the rise, affecting
patients with
cancer, organ transplant recipients, diabetics and those with HIV/AIDS. In
these patients,
invasive fungal infections may account for as many as 30% of deaths. Despite
the
development of a number of new antifungal agents, amphotericin B formulated as
an IV
administered micelle and liposomal dispersion remains one of the most
effective agents in
the treatment of systemic fungal infections. In addition, a variety of
parenteral
formulation approaches have been studied for AmpB. While effective, the
limitations of
these parenteral formulations of amphotericin B are the safety issues
associated with
administration (infection of the indwelling catheter, patient chills and
shaking due to

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
RBC haemolysis, dose-dependent renal toxicity), feasibility of administration
of
parenteral products in remote locations and high drug cost.
The development of an effective and safe oral formulation of amphotericin B
that
would have significant applications in the treatment of disseminated fungal
infections and
would dramatically expand access to treatment of visceral leishmaniasis.
However, the
bioavailability of AmpB is negligible due to low aqueous solubility and
instability at the
low pH found in gastric fluid. Such limitations also apply to a variety of
other
therapeutic agents for which oral formulations are desirable.
A need exists for effective and safe oral formulations of amphotericin B as
well as
many other therapeutic agents that provide for enhanced bioavailability and/or
increased
stability of the therapeutic agent of interest the low pH found in gastric
fluid. The present
invention seeks to fulfill these needs and provides further related
advantages.
SUMMARY OF THE INVENTION
The present invention provides compositions for formulating therapeutic
agents,
therapeutic agent formulations based on the compositions, methods for
administering
therapeutic agents using the formulations, and methods for treating conditions
and
diseases using the formulations.
In one aspect, the invention provides an amphotericin B formulation,
comprising,
(a) amphotericin B;
(b) one or more fatty acid glycerol esters; and
(c)
one or more polyethylene oxide-containing phospholipids or one or more
polyethylene oxide-containing fatty acid esters.
In one embodiment, amphotericin B is present in the formulation in an amount
from about 0.5 to about 10 mg/mL of the formulation. In one embodiment,
amphotericin
B is present in the formulation in about 5 mg/mL. In another embodiment,
amphotericin
B is present in the formulation in about 7 mg/mL.
In one embodiment, the fatty acid glycerol esters comprise from about 32 to
about
52% by weight fatty acid monoglycerides. In one embodiment, the fatty acid
glycerol
esters comprise from about 30 to about 50% by weight fatty acid diglycerides.
In one
embodiment, the fatty acid glycerol esters comprise from about 5 to about 20%
by weight
fatty acid triglycerides. In one embodiment, the fatty acid glycerol esters
comprise
greater than about 60% by weight oleic acid mono-, di-, and triglycerides.
-2-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
In one embodiment, the polyethylene oxide-containing phospholipids comprise a
C8-C22 saturated fatty acid ester of a phosphatidyl ethanolamine polyethylene
glycol salt.
In one embodiment, the polyethylene oxide-containing phospholipids comprise a
distearoylphosphatidyl ethanolamine polyethylene glycol salt. In one
embodiment, the
distearoylphosphatidyl ethanolamine polyethylene glycol salt is selected from
the group
consisting of a distearoylphosphatidyl ethanolamine polyethylene glycol 350
salt, a
distearoylphosphatidyl ethanolamine polyethylene glycol 550 salt, a
distearoylphosphatidyl ethanolamine polyethylene glycol 750 salt, a
distearoylphosphatidyl ethanolamine polyethylene glycol 1000 salt, a
distearoylphosphatidyl ethanolamine polyethylene glycol 2000 salt, and
mixtures thereof.
In one embodiment, the distearoylphosphatidyl ethanolamine polyethylene glycol
salt is
present in the formulation in an amount from 1 mM to about 30 mM based on the
volume
of the formulation. In one embodiment, the distearoylphosphatidyl ethanolamine

polyethylene glycol salt is an ammonium salt or a sodium salt.
In one embodiment, the polyethylene oxide-containing fatty acid esters
comprise
a polyethylene oxide ester of a C8-C22 saturated fatty acid. In one
embodiment, the
polyethylene oxide-containing fatty acid esters comprise a polyethylene oxide
ester of a
C12-C18 saturated fatty acid. In one embodiment, the polyethylene oxide-
containing
fatty acid esters is selected from the group consisting of lauric acid esters,
palmitic acid
esters, stearic acid esters, and mixtures thereof. In one embodiment, the
polyethylene
oxide-containing fatty acid esters comprise a polyethylene oxide having an
average
molecular weight of from about 750 to about 2000.
In one embodiment, the ratio of the fatty acid glycerol esters to polyethylene

oxide-containing fatty acid esters is from about 20:80 to about 80:20 v/v. In
one
embodiment, the ratio of the fatty acid glycerol esters to polyethylene oxide-
containing
fatty acid esters is about 60:40 v/v.
In one embodiment, the formulation further comprises glycerol in an amount
less
than about 10% by weight.
In one embodiment, the formulation is a self-emulsifying drug delivery system.
In another aspect, the invention provides a method for administering
amphotericin
B, comprising administering an amphotericin B formulation of the invention to
a subject
in need thereof. In one embodiment, the formulation is administered orally. In
another
embodiment, the formulation is administered topically.
-3-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
In another aspect, the invention provides a method for treating an infectious
disease treatable by the administration of amphotericin B, comprising
administering to a
subject in need thereof a therapeutically effective amount of an amphotericin
B
formulation of the invention. In one embodiment, the formulation is
administered orally.
In another embodiment, the formulation is administered topically.
Diseases treatable by the formulations include fungal infections, visceral
leishmaniasis, cutaneous leishmaniasis, Chagas disease, Alzheimer's disease,
or Febrile
neutropenia. Fungal infections treatable by the formulations include
aspergillosis,
blastomycosis, candidiasis, coccidioidomycosis, crytococcosis, histoplasmosis,
mucormycosis, paracoccidioidomycosis, or sporotrichosis.
In another aspect, the invention provides a formulation for the delivery of a
therapeutic agent, comprising,
(a) a therapeutic agent;
(b) one or more fatty acid glycerol esters; and
(c) one or more polyethylene oxide-containing phospholipids or one or more
polyethylene oxide-containing fatty acid esters.
In one embodiment, the therapeutic agent is present in the formulation in an
amount from about 0.1 mg/mL to about 25 mg/mL of the formulation.
In certain embodiments, the therapeutic agent is selected from the group
consisting of anticancers, antibiotics, antiviral drugs, antimycotics, anti-
prions, anti-
amoebics, non-steroidal anti-inflammatory drugs, anti-allergics,
immunosuppressive
agents, coronary drugs, analgesics, local anesthetics, anxiolytics, sedatives,
hypnotics,
migraine relieving agents, drugs against motion sickness, and anti-emetics.
In certain embodiments, the therapeutic agent is selected from the group
consisting of tetracycline, doxycycline, oxytetracycline, chloramphenicol,
erythromycin,
acyclovir, idoxuridine, tromantadine, miconazole, ketoconazole, fluconazole,
itraconazole, econazole, griseofulvin, amphotericin B, nystatine,
metronidazole,
metronidazole benzoate, tinidazole, indomethacin, ibuprofen, piroxicam,
diclofenac,
disodium cromoglycate, nitroglycerin, isosorbide dinitrate, verapamile,
nifedipine,
diltiazem, digoxine, morphine, cyclosporins, buprenorphine, lidocaine,
diazepam,
nitrazepam, flurazepam, estazolam, flunitrazepam, triazolam, alprazolam,
midazolam,
temazepam lormetazepam, brotizolam, clobazam, clonazepam, lorazepam, oxazepam,

busiprone, sumatriptan, ergotamine derivatives, cinnarizine, anti-histamines,
ondansetron,
-4-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
tropisetron, granisetrone, metoclopramide, disulfiram, vitamin K, paclitaxel,
docetaxel,
camptothecin, SN38, cisplatin, and carboplatin.
In one embodiment, the formulation further comprises a second therapeutic
agent.
In one embodiment, the fatty acid glycerol esters comprise from about 32 to
about
52% by weight fatty acid monoglycerides. In one embodiment, the fatty acid
glycerol
esters comprise from about 30 to about 50% by weight fatty acid diglycerides.
In one
embodiment, the fatty acid glycerol esters comprise from about 5 to about 20%
by weight
fatty acid triglycerides. In one embodiment, the fatty acid glycerol esters
comprise
greater than about 60% by weight oleic acid mono-, di-, and triglycerides.
In one embodiment, the polyethylene oxide-containing phospholipids comprise a
C8-C22 saturated fatty acid ester of a phosphatidyl ethanolamine polyethylene
glycol salt.
In one embodiment, the polyethylene oxide-containing phospholipids comprise a
distearoylphosphatidyl ethanolamine polyethylene glycol salt. In one
embodiment, the
distearoylphosphatidyl ethanolamine polyethylene glycol salt is selected from
the group
consisting of a distearoylphosphatidyl ethanolamine polyethylene glycol 350
salt, a
distearoylphosphatidyl ethanolamine polyethylene glycol 550 salt, a
distearoylphosphatidyl ethanolamine polyethylene glycol 750 salt, a
distearoylphosphatidyl ethanolamine polyethylene glycol 1000 salt, a
distearoylphosphatidyl ethanolamine polyethylene glycol 2000 salt, and
mixtures thereof.
In one embodiment, the distearoylphosphatidyl ethanolamine polyethylene glycol
salt is
present in the formulation in an amount from 1 mM to about 30 mM based on the
volume
of the formulation. In one embodiment, the distearoylphosphatidyl ethanolamine

polyethylene glycol salt is an ammonium salt or a sodium salt.
In one embodiment, the polyethylene oxide-containing fatty acid esters
comprise
a polyethylene oxide ester of a C8-C22 saturated fatty acid. In one
embodiment, the
polyethylene oxide-containing fatty acid esters comprise a polyethylene oxide
ester of a
Cl 2-C18 saturated fatty acid. In one embodiment, the polyethylene oxide-
containing
fatty acid esters is selected from the group consisting of lauric acid esters,
palmitic acid
esters, stearic acid esters, and mixtures thereof. In one embodiment, the
polyethylene
oxide-containing fatty acid esters comprise a polyethylene oxide having an
average
molecular weight of from about 750 to about 2000.
In one embodiment, the ratio of the fatty acid glycerol esters to polyethylene

oxide-containing fatty acid esters is from about 20:80 to about 80:20 v/v. In
one
-5-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
embodiment, the ratio of the fatty acid glycerol esters to polyethylene oxide-
containing
fatty acid esters is about 60:40 v/v.
In one embodiment, the formulation further comprises glycerol in an amount
less
than about 10% by weight.
In one embodiment, the formulation is a self-emulsifying drug delivery system.
In another aspect, the invention provides a method for administering a
therapeutic
agent, comprising administering a therapeutic agent formulation of the
invention to a
subject in need of such agent. In one embodiment, the formulation is
administered orally.
In another embodiment, the formulation is administered topically.
In another aspect, the invention provides a composition for formulating a
therapeutic agent, comprising,
(a) one or more fatty acid glycerol esters; and
(b) one or more polyethylene oxide-containing phospholipids or one or more
polyethylene oxide-containing fatty acid esters.
In one embodiment, the fatty acid glycerol esters comprise from about 32 to
about
52% by weight fatty acid monoglycerides. In one embodiment, the fatty acid
glycerol
esters comprise from about 30 to about 50% by weight fatty acid diglycerides.
In one
embodiment, the fatty acid glycerol esters comprise from about 5 to about 20%
by weight
fatty acid triglycerides. In one embodiment, the fatty acid glycerol esters
comprise
greater than about 60% by weight oleic acid mono-, di-, and triglycerides.
In one embodiment, the polyethylene oxide-containing phospholipids comprise a
C8-C22 saturated fatty acid ester of a phosphatidyl ethanolamine polyethylene
glycol salt.
In one embodiment, the polyethylene oxide-containing phospholipids comprise a
distearoylphosphatidyl ethanolamine polyethylene glycol salt. In one
embodiment, the
distearoylphosphatidyl ethanolamine polyethylene glycol salt is selected from
the group
consisting of a distearoylphosphatidyl ethanolamine polyethylene glycol 350
salt, a
distearoylphosphatidyl ethanolamine polyethylene glycol 550 salt, a
distearoylphosphatidyl ethanolamine polyethylene glycol 750 salt, a
distearoylphosphatidyl ethanolamine polyethylene glycol 1000 salt, a
distearoylphosphatidyl ethanolamine polyethylene glycol 2000 salt, and
mixtures thereof.
In one embodiment, the distearoylphosphatidyl ethanolamine polyethylene glycol
salt is
present in the formulation in an amount from 1 mM to about 30 mM based on the
volume
-6-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
of the formulation. In one embodiment, the distearoylphosphatidyl ethanolamine

polyethylene glycol salt is an ammonium salt or a sodium salt.
In one embodiment, the polyethylene oxide-containing fatty acid esters
comprise
a polyethylene oxide ester of a C8-C22 saturated fatty acid. In one
embodiment, the
polyethylene oxide-containing fatty acid esters comprise a polyethylene oxide
ester of a
C 12-C18 saturated fatty acid. In one embodiment, the polyethylene oxide-
containing
fatty acid esters is selected from the group consisting of lauric acid esters,
palmitic acid
esters, stearic acid esters, and mixtures thereof. In one embodiment, the
polyethylene
oxide-containing fatty acid esters comprise a polyethylene oxide having an
average
molecular weight of from about 750 to about 2000.
In one embodiment, the composition further comprises glycerol in an amount
less
than about 10% by weight.
In another aspect, the invention provides a method for formulating a
therapeutic
agent, comprising combining a therapeutic agent with a composition of the
invention for
formulating a therapeutic agent.
DESCRIPTION OF THE DRAWINGS
The foregoing aspects and many of the attendant advantages of this invention
will
become more readily appreciated as the same become better understood by
reference to
the following detailed description, when taken in conjunction with the
accompanying
drawings.
FIGURE 1A illustrates the chemical structure of amphotericin B (AmpB).
FIGURE 1B illustrates the chemical structure of distearoylphosphatidyl
ethanolamine polyethylene glycol 2000 ammonium salt (DSPE-PEG-2000).
FIGURE 2 compares AmpB concentration ( g/mL) in an AmpB/PECEOL
formulation and representative AmpB formulations of the invention
(AmpB/PECEOC/DSPE-PEG-2000) containing DSPE-PEG-2000 at concentrations of
5, 10, and 15mM.
FIGURE 3A compares the UV absorbance spectra over time of representative
AmpB formulations of the invention (PECEOLc)/DSPE-PEG) at various
concentrations
(0.5-15 g/ml) incubated in simulated gastric fluid (SGF).
FIGURE 3B compares standard curves from the data in FIGURE 2A combined
using peak height at 407 nm to construct the standard curves of AmpB
absorbance vs.
-7-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
concentration. A different standard curve was prepared for each formulation
where
DSPE-PEG molecular weight varied.
FIGURE 4 compares the stability of AmpB in representative formulations of the
invention (PECEOL /DSPE-PEG 350, 550, 750, and 2000) with an AmpB/PECEOL
formulation at 37 C in simulated gastric fluid as a function of time (10, 30,
and 120 min).
FIGURES 5A and 5B compare the stability of AmpB in representative
formulations of the invention (PECEOL /DSPE-PEG 350, 550, 750, and 2000,
designated PEG 350, 550, 750, 2000, respectively) with an AmpB/PECEOL
formulation
at 37 C in fasted-state simulated intestinal fluid (FSSIF) without lecithin
(5A) and with
lecithin (5B) as a function of time (10, 30, 60, and 120 min).
FIGURE 6 compares the stability of AmpB in representative formulations of the
invention (PECEOL /DSPE-PEG 350, 550, 750, and 2000, designated PEG 350, 550,
750, 2000, respectively) with an AmpB/PECEOL formulation at 37 C in simulated

intestinal fluid (S1F) with pancreatin enzymes as a function of time (10, 30,
60, and
120 min).
FIGURE 7 compares Candida albicans concentration (CFU/ml) in the kidneys of
rats infected with Candida albicans and treated with control, an AmpB/PECEOL
formulation (10mg/kg), a representative AmpB formulation of the invention
(AmpB/PECEOL /DSPE-PEG-2000, designated AmpB/DSPE-PEG-2000, 10mg/kg),
and intravenous ABELCET (designated ABLC, 5mg/m1).
FIGURE 8 compares Candida albicans concentration (CFU/ml) in the organs of
rats infected with Candida albicans and treated with control, an AmpB/PECEOL
formulation (10mg/kg), a representative AmpB formulation of the invention
(AmpB/PECEOL /DSPE-PEG-2000, designated AmpB/DSPE-PEG-2000, 10mg/kg),
and intravenous ABELCET (designated ABLC, 5mg/m1).
FIGURE 9 compares plasma creatinine (mg/de in rats infected with Candida
albicans and treated with control, an AmpB/PECEOL formulation (10mg/kg), a
representative AmpB formulation of the invention (AmpB/PECEOL /DSPE-PEG-2000,
designated AmpB/DSPE-PEG-2000, 10mg/kg), and intravenous ABELCET (designated
ABLC, 5mg/m1) (blank, Ohr, and 48hr).
FIGURES 10A, 10B, and 10C compare AmpB concentration (mg/mL) in
representative AmpB formulations of the invention (AmpB/PECEOL /GELUCIRE
44/14; AmpB/PECEOL /GELUCIRE 50/13; and AmpB/PECEOC/GELUCIRE
-8-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
53/10) at varying ratios of PECEOL :GELUC1RE (60:40; 50:50; and 40:60 v/v) at
2, 4
and 24hrs.
FIGURE 11 compares AmpB concentration (% original concentration, 5mg/mL)
over time (0, 1, 5, 7, 15, 21, 28, 36, 43, 49, and 56 days) for an AmpB/PECEOL

formulation (designated PECEOL ) and representative AmpB formulations of the
invention (AmpB/PECEOL GELUC1RE 44/14, 50/50; and AmpB/PECEOC/DSPE-
PEG-2000, 15 mM DSPE-PEG-2000).
FIGURE 12 compares AmpB concentration (% original concentration, 5mg/mL)
over time (10, 30, 45, 60, 90, and 120min) in simulated gastric fluid (SGF)
for an
AmpB/PECEOL formulation and representative AmpB formulations of the invention
(AmpB/PECEOL /GELUC1RE 44/14, 50/50; and AmpB/PECEOL /DSPE-PEG-2000,
mM DSPE-PEG-2000).
FIGURE 13 compares AmpB concentration (% original concentration, 5mg/mL)
over time (10, 30, 45, 60, 90, 120, and 240min) in fed-state simulated
intestinal fluid
15
(FeSSIF) for an AmpB/PECEOL formulation and a representative AmpB formulation
of
the invention (AmpB/PECEOL /GELUCIRE 44/14, 50/50).
FIGURE 14 compares AmpB concentration (% original concentration, 5mg/mL)
over time (10, 30, 45, 60, 90, 120, and 240min) in fed-state simulated
intestinal fluid
(FeSSW) with enzyme for an AmpB/PECEOL formulation and a representative AmpB
formulation of the invention (AmpB/PECEOL GELUCIRE 44/14, 50/50).
FIGURE 15 compares AmpB concentration (% original concentration, 5mg/mL)
over time (10, 30, 45, 60, 90, 120, and 240min) in fasted-state simulated
intestinal fluid
(FaSSLF) for an AmpB/PECEOL formulation and a representative AmpB formulation
of
the invention (AmpB/PECEOL GELUCIRE 44/14, 50/50).
FIGURE 16 compares AmpB concentration (% original concentration, 10mg/mL)
of a representative AmpB formulation of the invention (AmpB/PECEOC/DSPE-
PEG-2000, 15 mM DSPE-PEG-2000) after seven days at room temperature and 43 C
(AmpB measured by UV absorbance of centrifuged samples after specified time).
DETAILED DESCRIPTION OF THE INVENTION
The present invention provides compositions for formulating therapeutic
agents.
The compositions are effective for solubilizing therapeutic agents,
particularly difficultly
soluble therapeutic agents. The compositions advantageously enhance the
bioavailability
of the therapeutic agents. The invention also provides therapeutic agent
formulations
-9-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
based on the compositions that are effective for the delivery of therapeutic
agents,
particularly oral administration of therapeutic agents. Amphotericin B
formulations are
used herein as the prototypic example, however, one of skill in the art will
appreciate that
such formulations are applicable to a variety of therapeutic agents.
Accordingly, in one
aspect, the invention provides amphotericin B formulations based on the
compositions.
The amphotericin B formulations effectively solubilize amphotericin B
providing
formulations having increased amphotericin B concentrations and, at the same
time,
provide for enhanced amphotericin B bioavailability.
Amphotericin B Formulations
In one aspect, the present invention provides amphotericin B formulations,
methods for making the formulations, methods for administering amphotericin B
using
the formulations, and methods for treating diseases treatable by amphotericin
B by
administering the formulations.
Amphotericin B is an effective antifungal agent, and at present, is the drug
of
choice for treating most serious systemic fungal infections. The drug binds
strongly to
ergosterol, a major sterol component of fungal membranes, forming pores in the

membranes causing disruption of the membrane, cell permeability, and lysis.
Amphotericin B has had limitations in clinical administration due to several
unfavorable properties.
First, amphotericin B has a strong binding affinity for
cholesterol, a sterol present in most mammalian cell membranes, and therefore
is capable
of disrupting host cells. This leads to renal toxicity of the drug. Second,
amphotericin B
is not absorbed in the gastrointestinal tract (GIT) due to its poor solubility
and its
sensitivity to the acid environment of the stomach. To overcome this problem,
amphotericin B is used parenterally as liposomal (AMBISOME ) or as colloidal
dispersion (FUNGIZONE , ABELCET ) for the treatment of certain systemic fungal
infections (Arikan and Rex, 2001. Lipid-based antifungal agents: current
status. Curr.
Phann. Des. 5, 393-415).
However, intravenous injection and infusion of amphotericin B have significant

disadvantages. First, the intravenous injection and infusion of amphotericin B
has been
associated with considerable fluctuation of drug concentrations in the blood
and side
effects such as nephrotoxicity (Mailer et al., 2000, Nanosuspensions for the
formulation
of poorly soluble drugs-rationale for development and what we can expect for
the future.
In: Nielloud, F., Marti-Mestres, G. (Eds.), Pharmaceutical emulsions and
suspensions.
-10-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
Plenum Press/Marcel Dekker, New York, pp. 383-408). Second, in addition to the
high
cost, the injection and infusion formulation of amphotericin B have also
presented low
compliance and technical problems with administration in endemic countries.
In one embodiment, the present invention overcomes these disadvantages by
providing an amphotericin B formulation that can be administered orally. The
oral
amphotericin B formulations of the invention can be expected to improve
patient
compliance and to improve pharmacokinetics of the drug and to increase the
amphotericin B absorption in GI track.
Amphotericin B is an antimycotic polyene antibiotic obtained from Streptomyces
nodosus M4575.
Amphotericin B is designated chemically as
[1R-(1R*,3S*,5R*,6R*,9R*,11R*,15S*,16R*,17R*,18S*,19E,21E,23E,25E, 27E,29E,31
E,33R*,35S*,36R*,37S 0]-33- [(3- amino-3,6-dideoxy-B-D-mannopyranosyl)
oxy] 1,3,5,6,9,11,17,37-octahydroxy-15,16,18-trimethy1-13-oxo-14,39-diox
abicyclo-
[33 .3.1] nonatriaconta-19,21,23,25,27,29,31-heptaene-36-carboxylic acid. The
chemical
structure of amphotericin B is shown in FIGURE 1A. Crystalline amphotericin B
is
insoluble in water.
In one aspect, the present invention provides amphotericin B formulations. The

amphotericin formulations of the invention include
(a) amphotericin B;
(b) one or more fatty acid glycerol esters; and
(c)
one or more polyethylene oxide-containing phospholipids or one or more
polyethylene oxide-containing fatty acid esters.
In representative formulations, amphotericin B is present in an amount from
about
0.5 to about 10 mg/mL of the formulation. In one embodiment, amphotericin B or
pharmaceutically acceptable salt thereof is present in the formulation in
about 5 mg/mL.
In one embodiment, amphotericin B or its pharmaceutically acceptable salt
thereof is
present in the formulation in about 7 mg/mL.
The amphotericin B formulations include one or more fatty acid glycerol
esters,
and typically, a mixture of fatty acid glycerol esters. As used herein the
term "fatty acid
glycerol esters" refers to esters formed between glycerol and one or more
fatty acids
including mono-, di-, and tri-esters (i.e., glycerides). Suitable fatty acids
include
saturated and unsaturated fatty acids having from eight (8) to twenty-two (22)
carbons
-11-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
atoms (i.e., C8-C22 fatty acids). In certain embodiments, suitable fatty acids
include
C12-C18 fatty acids.
The fatty acid glycerol esters useful in the formulations can be provided by
commercially available sources. A representative source for the fatty acid
glycerol esters
is a mixture of mono-, di-, and triesters commercially available as PECEOL
(Gattefosse,
Saint Priest Cedex, France), commonly referred to as "glyceryl oleate" or
"glyceryl
monooleate." When PECEOL is used as the source of fatty acid glycerol esters
in the
formulations, the fatty acid glycerol esters comprise from about 32 to about
52% by
weight fatty acid monoglycerides, from about 30 to about 50% by weight fatty
acid
diglycerides, and from about 5 to about 20% by weight fatty acid
triglycerides. The fatty
acid glycerol esters comprise greater than about 60% by weight oleic acid
(C18:1) mono-,
di-, and triglycerides. Other fatty acid glycerol esters include esters of
palmitic acid
(C16) (less than about 12%), stearic acid (C18) (less than about 6%), linoleic
acid
(C18:2) (less than about 35%), linolenic aid (C18:3) (less than about 2%),
arachidie acid
(C20) (less than about 2%), and eicosenoic acid (C20:1) (less than about 2%).
PECEOL
can also include free glycerol (typically about 1%). In one embodiment, the
fatty acid
glycerol esters comprise about 44% by weight fatty acid monoglycerides, about
45% by
weight fatty acid diglycerides, and about 9% by weight fatty acid
triglycerides, and the
fatty acid glycerol esters comprise about 78% by weight oleic acid (C18:1)
mono-, di-,
and triglycerides. Other fatty acid glycerol esters include esters of palmitic
acid (C16)
(about 4%), stearic acid (C18) (about 2%), linoleic acid (C18:2) (about 12%),
linolenic
aid (C18:3) (less than 1%), arachidic acid (C20) (less than 1%), and
eicosenoic acid
(C20:1) (less than 1%).
In certain embodiments, the formulations of the invention can include glycerol
in
an amount less than about 10% by weight.
Amphotericin B Formulations: Polyethylene Oxide-Containing Phospholipids
(DSPE-PEGs). The amphotericin B formulations include one or more
polyethoxylated
lipids. In one embodiment, the polyethoxylated lipids are polyethylene oxide-
containing
phospholipids, or a mixture of polyethylene oxide-containing phospholipids. In
another
embodiment, the polyethoxylated lipids are polyethylene oxide-containing fatty
acid
esters, or a mixture of polyethylene oxide-containing fatty acid esters.
Accordingly, in one embodiment, the amphotericin B formulations of the
invention include
-12-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
(a) amphotericin B;
(b) one or more fatty acid glycerol esters; and
(c) one or more polyethylene oxide-containing phospholipids.
As used herein, the term "polyethylene oxide-containing phospholipid" refers
to a
phospholipid that includes a polyethylene oxide group (i.e., polyethylene
glycol group)
covalently coupled to the phospholipid, typically through a carbamate or an
ester bond.
Phospholipids are derived from glycerol and can include a phosphate ester
group and two
fatty acid ester groups. Suitable fatty acids include saturated and
unsaturated fatty acids
having from eight (8) to twenty-two (22) carbons atoms (i.e., C8-C22 fatty
acids). In
certain embodiments, suitable fatty acids include saturated C12-C18 fatty
acids.
Representative polyethylene oxide-containing phospholipids include C8-C22
saturated
fatty acid esters of a phosphatidyl ethanolamine polyethylene glycol salt. In
certain
embodiments, suitable fatty acids include saturated C12-C18 fatty acids.
The molecular weight of the polyethylene oxide group of the polyethylene
oxide-containing phospholipid can be varied to optimize the solubility of the
therapeutic
agent (e.g., amphotericin B) in the formulation. Representative average
molecular
weights for the polyethylene oxide groups can be from about 200 to about 5000
(e.g., PEG 200 to PEG 5000).
In one embodiment, the polyethylene oxide-containing phospholipids are
distearoyl phosphatidyl ethanolamine polyethylene glycol salts.
Representative
distearoylphosphatidyl ethanolamine polyethylene glycol
salts include
distearoylphosphatidyl ethanolamine polyethylene glycol 350 (DSPE-PEG-350)
salts,
distearoylphosphatidyl ethanolamine polyethylene glycol 550 (DSPE-PEG-550)
salts,
distearoylphosphatidyl ethanolamine polyethylene glycol 750 (DSPE-PEG-750)
salts,
distearoylphosphatidyl ethanolamine polyethylene glycol 1000 (DSPE-PEG-1000)
salts,
distearoylphosphatidyl ethanolamine polyethylene glycol 1500 (DSPE-PEG-1500)
salts,
and distearoylphosphatidyl ethanolamine polyethylene glycol 2000 (DSPE-PEG-
2000)
salts. Mixtures can also be used. For the distearoylphosphatidyl ethanolamine
polyethylene glycol salts above, the number (e.g., 350, 550, 750, 1000, and
2000)
designates the average molecular weight of the polyethylene oxide group. The
abbreviations for these salts used herein is provided in parentheses above.
Suitable distearoylphosphatidyl ethanolamine polyethylene glycol salts include

ammonium and sodium salts.
-13-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
The chemical structure of distearoylphosphatidyl ethanolamine polyethylene
glycol 2000 (DSPE-PEG-2000) ammonium salt is illustrated in FIGURE 1B.
Referring
to FIGURE 1B, the polyethylene oxide-containing phospholipid includes a
phosphate
ester group and two fatty acid ester (stearate) groups, and a polyethylene
oxide group
covalently coupled to the amino group of the phosphatidyl ethanolamine through
a
carbamate bond.
As noted above, the polyethylene oxide-containing phospholipid affects the
ability
of the formulation to solubilize a therapeutic agent. In general, the greater
the amount of
polyethylene oxide-containing phospholipid, the greater the solubilizing
capacity of the
formulation for difficultly soluble therapeutic agents. The polyethylene oxide-
containing
phospholipid can be present in the formulation in an amount from about 1 mM to
about
30 mM based on the volume of the formulation. In certain embodiments, the
distearoylphosphatidyl ethanolamine polyethylene glycol salt is present in the

formulation in an amount from 1 mM to about 30 mM based on the volume of the
formulation. In one embodiment, the distearoylphosphatidyl ethanolamine
polyethylene
glycol salt is present in the formulation in about 15 mM based on the volume
of the
formulation.
FIGURE 2 compares amphotericin B concentration (p.g/mL) in an
AmpB/PECEOL formulation (containing no polyethylene oxide-containing
phospholipids or polyethylene oxide-containing fatty acid esters) and
representative
AmpB formulations of the invention (AmpB/PECEOL /DSPE-PEG-2000) containing
DSPE-PEG-2000 at concentrations of 5, 10, and 15mM. AmpB measured by UV
absorbance of centrifuged samples after 24hrs at 45 C.
In one embodiment, the amphotericin B formulations of the invention include
(a) amphotericin B;
(b) oleic acid glycerol esters; and
(c) a distearoylphosphatidyl ethanolamine polyethylene glycol salt.
In one embodiment, the amphotericin B formulation of the invention includes
amphotericin B, PECEOL , and a distearoylphosphatidyl ethanolamine
polyethylene
glycol salt. In this embodiment, the distearoylphosphatidyl ethanolamine
polyethylene
glycol salt is present in an amount up to about 30 mM.
-14-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
The preparation and characterization of representative amphotericin B
formulations of the invention that include polyethylene oxide-containing
phospholipids is
described in Example 1.
The amphotericin B formulations that include polyethylene oxide-containing
phospholipids include amphotericin B that is both partially solubilized
(dissolved) and
present as solid particles to provide a fine solid dispersion. Dispersion of
the formulation
in aqueous media provides a nano-/microemulsion having emulsion droplets that
range in
size from about 50 nm to about 5 pm.
Polyethylene glycol molecular weight had no clear effect on the emulsion
droplet
size in simulated intestinal fluid (Table 3) following mixing over a period of
2h at 37 C.
Submicron mean diameters were observed in the range of 300-600nm with a fairly
wide
polydispersity. A bimodal particle size distribution was also generated, with
a small
subpopulation (about 20%) centered in submicron range (150-300nm) and another
centered in the 1-2pm range (about 80%). AmpB in PECEOL alone also formed
droplets of similar size and distribution in simulated intestinal fluid.
To determine their effectiveness as orally administered formulations, the
stability
of representative amphotericin B formulations of the invention was evaluated
in
simulated gastric fluid. FIGURE 3A compares the UV absorbance spectra over
time of
representative AmpB formulations of the invention (PECEOC/DSPE-PEG) at various
concentrations (0.5-15 g/ml) incubated in simulated gastric fluid (SGF).
There is no
change in the peak height or peak ratio at any concentration as a function of
incubation
time up to 60 min. FIGURE 3B compares standard curves from the data in FIGURE
2A
combined using peak height at 407 nm to construct the standard curves of AmpB
absorbance vs. concentration. A different standard curve was prepared for each
formulation where DSPE-PEG molecular weight varied. FIGURE 4 compares the
stability of AmpB in representative formulations of the invention (PECEOC/DSPE-
PEG
350, 550, 750, and 2000) with an AmpB/PECEOL formulation at 37 C in simulated

gastric fluid as a function of time (10, 30, and 120 min). Data represent the
mean SD of
three independent experiments, each of which was performed in triplicate. Each
of the
evaluated representative formulations of the invention demonstrated stability
in simulated
gastric fluid over the time period evaluated.
The stability of representative amphotericin B formulations of the invention
was
also evaluated in fasted-state simulated intestinal fluid (FSSIF) without
lecithin and with
-15-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
lecithin, and in simulated intestinal fluid with pancreatin enzymes. FIGURES
5A and 5B
compare the stability of AmpB in representative formulations of the invention
(PECE012/DSPE-PEG 350, 550, 750, and 2000, designated PEG 350, 550, 750, 2000,

respectively) with an AmpB/PECEOL formulation at 37 C in fasted-state
simulated
intestinal fluid (FSSIF) without lecithin (5A) and with lecithin (5B) as a
function of time
(10, 30, 60, and 120 min). Data represent the mean SD of three independent
experiments, each of which was performed in triplicate. FIGURE 6 compares the
stability of AmpB in representative formulations of the invention (PECEOL
/DSPE-PEG
350, 550, 750, and 2000, designated PEG 350, 550, 750, 2000, respectively)
with an
AmpB/PECEOL formulation at 37 C in simulated intestinal fluid (SIF) with
pancreatin
enzymes as a function of time (10, 30, 60, and 120 min) Data represent the
mean SD of
three independent experiments, each of which was performed in triplicate. Each
of the
evaluated representative formulations of the invention demonstrated stability
in the
simulated intestinal fluids over the time period evaluated.
The stability of the representative amphotericin B formulations in the GI
fluids
demonstrates their suitability as orally administered formulations.
AmpB in PECEOL was stabilized and its solubility enhanced 50-fold by the
incorporation of 15mM DSPE-PEG, where the PEG mean molecular weight was varied

between 350 and 2000. Drug stability in stomach and intestine is critical for
promoting
drug absorption in the GI tract. AmpB is well known to be more soluble but
relatively
unstable at low pH, therefore any protection afforded by the lipid components
of the
formulation could be a significant benefit toward increasing the oral
bioavailability of
AmpB. It
is also important to know if the lipidic vehicles influenced the
superaggregation state of AmpB, which has been previously been shown to
influence
drug solubility as well as in vivo activity. The UV spectral pattern of AmpB
in the lipidic
vehicles described herein was consistent with monomeric AmpB before and after
incubation in simulated gastric or intestinal fluids (see FIGURE 3A). No UV
spectral
pattern change was noted upon ambient temperature storage (21 C) over a period
of
4 weeks either (data not shown). However, interactions between the AmpB and
the lipid
components in the undiluted formulation (in the absence of the assay solvent)
or
following oral absorption in vivo may be different.
Stability of representative formulations of the invention in simulated gastric
fluid
over 2h was excellent, with surprisingly little variability between
formulations prepared
-16-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
with the various DSPE-PEGs or with only PECEOL (see FIGURES 4-6). All showed
a
translucent appearance with no precipitate appearing. More variation in
stability was
observed in simulated fasted-state intestinal fluid containing bile salts (see
FIGURES 5A
and 5B). The emulsification properties of the bile salts, lecithin and
phospholipase in
pancreatin could influence formulation stability and therefore drug stability
was evaluated
in simulated intestinal fluids containing these components. Lecithin would
likely be
incorporated into the lipid mixture when including in the simulated intestinal
fluid, which
had the potential to either improve the association of amphotericin B with the
lipid
excipients or to exclude it. The presence of lecithin, however, made no
appreciable
difference in the rate or extent of degradation or in the rank order of
degradation at the
end of 2h (see FIGURE 5B). Clearly, DSPE-PEG 350 containing formulations
provided
less drug stability than those containing the longer-chain PEGs. In the
absence of
lecithin, submicron particle size analysis did not show a significant
population below
50nm that would be consistent with DSPE-PEG micelles, e.g., if DSPE-PEG350 had
self-associated into a separate micelle population (see FIGURE 5A).
Furthermore, there
was no significant effect on particle size due to the presence of DSPE-PEG or
polyethylene glycol molecular weight, suggesting that the emulsification
properties are
largely derived from the PECEOL component. However, the possibility that a
small
fraction of micelles exists in equilibrium with the lipid excipient/drug
mixture in the
simulated intestinal fluid cannot be excluded for any of the DSPE-PEG
formulations,
however, it does not appear to be a major component. Therefore, it is possible
that the
improved stability of AmpB in PECEOL /DSPE-PEG of higher molecular weight may
be related to the surface properties of the emulsion droplets themselves, in
spite of the
lack of a direct relationship to particle size distribution, such that the
hydrophilic
polyethylene glycol chains may orient to the water interface while the PECEOL
/AmpB
fraction would remain in the inner oil phase and thereby sequestering and
protecting the
AmpB from degradation. Thus, there was a trend to increased stability for PEG
molecular weight 750 and 2000 compared to 350 and 550. The interactions
between
AmpB and the PECEOL /DSPE-PEG resulted in a UV spectral shape consistent with
monomeric AmpB rather than aggregated AmpB (FIGURE 3A).
Amphotericin B Formulations: Polyethylene Oxide-Containing Fatty Acid Esters.
As noted above, the amphotericin B formulations include one or more
polyethoxylated
lipids such as polyethylene oxide-containing phospholipids or one or more
polyethylene
-17-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
oxide-containing fatty acid esters, and typically, a mixture of polyethylene
oxide-containing phospholipids or a mixture of polyethylene oxide-containing
fatty acid
esters.
Accordingly, in one embodiment, the amphotericin B formulations of the
invention include
(a) amphotericin B;
(b) one or more fatty acid glycerol esters; and
(c) one or more polyethylene oxide-containing fatty acid esters.
As used herein, the term "polyethylene oxide-containing fatty acid ester"
refers to
a fatty acid ester that includes a polyethylene oxide group (i.e.,
polyethylene glycol
group) covalently coupled to the fatty acid through an ester bond.
Polyethylene
oxide-containing fatty acid esters include mono- and di-fatty acid esters of
polyethylene
glycol. Suitable polyethylene oxide-containing fatty acid esters are derived
from fatty
acids including saturated and unsaturated fatty acids having from eight (8) to
twenty-two
(22) carbons atoms (i.e., a polyethylene oxide ester of a C8-C22 fatty acid).
In certain
embodiments, suitable polyethylene oxide-containing fatty acid esters are
derived from
fatty acids including saturated and unsaturated fatty acids having from twelve
(12) to
eighteen (18) carbons atoms (i.e., a polyethylene oxide ester of a C12-C18
fatty acid).
Representative polyethylene oxide-containing fatty acid esters include
saturated C8-C22
fatty acid esters. In certain embodiments, suitable polyethylene oxide-
containing fatty
acid esters include saturated C12-C18 fatty acids.
The molecular weight of the polyethylene oxide group of the polyethylene oxide-

containing fatty acid ester can be varied to optimize the solubility of the
therapeutic agent
(e.g., amphotericin B) in the formulation. Representative average molecular
weights for
the polyethylene oxide groups can be from about 350 to about 2000. In one
embodiment,
the average molecular weight for the polyethylene oxide group is about 1500.
In this embodiment, the amphotericin B formulations include one or more
polyethylene oxide-containing fatty acid esters, and typically, a mixture of
polyethylene
oxide-containing fatty acid esters (mono- and di-fatty acid esters of
polyethylene glycol).
The polyethylene oxide-containing fatty acid esters useful in the formulations
can
be provided by commercially available sources. Representative polyethylene
oxide-
containing fatty acid esters (mixtures of mono- and diesters) are commercially
available
under the designation GELUCIRE (Gattefosse, Saint Priest Cedex, France).
Suitable
-18-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
polyethylene oxide-containing fatty acid esters can be provided by GELUCIRE
44/14,
GELUCIRE 50/13, and GELUCIRE 53/10. The numerals in these designations refer

to the melting point and hydrophilic/lipophilic balance (HLB) of these
materials,
respectively.
GELUCIRE 44/14, GELUCIRE 50/13, and GELUCIRE 53/10 are mixtures of
(a) mono-, di-, and triesters of glycerol (glycerides) and (b) mono- and
diesters of
polyethylene glycol (macrogols). The GELUCIRES can also include free
polyethylene
glycol (e.g., PEG 1500).
Lauric acid (C12) is the predominant fatty acid component of the glycerides
and
polyethylene glycol esters in GELUCIRE 44/14. GELUCIRE 44/14 is referred to
as a
mixture of glyceryl dilaurate (lauric acid diester with glycerol) and PEG
dilaurate (lauric
acid diester with polyethylene glycol), and is commonly known as PEG-32
glyceryl
laurate (Gattefosse) lauroyl macrogo1-32 glycerides EP, or lauroyl
polyoxylglycerides
USP/NF. GELUCIRE 44/14 is produced by the reaction of hydrogenated palm
kernel
oil with polyethylene glycol (average molecular weight 1500). GELUCIRE 44/14
includes about 20% mono-, di- and, triglycerides, about 72% mono- and di-fatty
acid
esters of polyethylene glycol 1500, and about 8% polyethylene glycol 1500.
GELUCIRE 44/14 includes lauric acid (C12) esters (30 to 50%), myristic acid
(C14) esters (5 to 25%), palmitic acid (C16) esters (4 to 25%), stearic acid
(C18) esters
(5 to 35%), caprylic acid (C8) esters (less than 15%), and capric acid (C10)
esters (less
than 12%). GELUCIRE 44/14 may also include free glycerol (typically less than
about
1%). In a representative formulation, GELUCIRE 44/14 includes lauric acid
(C12)
esters (about 47%), myristic acid (C14) esters (about 18%), palmitic acid
(C16) esters
(about 10%), stearic acid (C18) esters (about 11%), caprylic acid (C8) esters
(about 8%),
and capric acid (C10) esters (about 12%).
Palmitic acid (C16) (40-50%) and stearic acid (C18) (48-58%) are the
predominant fatty acid components of the glycerides and polyethylene glycol
esters in
GELUCIRE 50/13. GELUCIRE 50/13 is known as PEG-32 glyceryl palmitostearate
(Gattefosse), stearoyl macrogolglycerides EP, or stearoyl polyoxylglycerides
USP/NF).
GELUCERE 50/13 includes palmitic acid (C16) esters (40 to 50%), stearic acid
(C18)
esters (48 to 58%) (stearic and palmitic acid esters greater than about 90%),
lauric acid
(C12) esters (less than 5%), myristic acid (C14) esters (less than 5%),
caprylic acid (C8)
esters (less than 3%), and capric acid (C10) esters (less than 3%). GELUCIRE
50/13
-19-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
may also include free glycerol (typically less than about 1%). In a
representative
formulation, GELUCIRE 50/13 includes palmitic acid (C16) esters (about 43%),
stearic
acid (C18) esters (about 54%) (stearic and palmitic acid esters about 97%),
lauric acid
(C12) esters (less than 1%), myristic acid (C14) esters (about 1%), caprylic
acid (C8)
esters (less than 1%), and capric acid (C10) esters (less than 1%)
Stearic acid (C18) is the predominant fatty acid component of the glycerides
and
polyethylene glycol esters in GELUCIRE 53/10. GELUCIRE 53/10 is known as
PEG-32 glyceryl stearate (Gattefosse).
In one embodiment, the polyethylene oxide-containing fatty acid ester is a
lauric
acid ester, a palmitic acid ester, or a stearic acid ester (i.e., mono- and di-
lauric acid esters
of polyethylene glycol, mono- and di-palmitic acid esters of polyethylene
glycol,
mono- and di-stearic acid esters of polyethylene glycol). Mixtures of these
esters can
also be used.
For embodiments that include polyethylene oxide-containing fatty acid esters,
the
ratio of the fatty acid glycerol esters to polyethylene oxide-containing fatty
acid esters is
from about 20:80 to about 80:20 v/v. In one embodiment, the ratio of the fatty
acid
glycerol esters to polyethylene oxide-containing fatty acid esters is about
30:70 v/v. In
one embodiment, the ratio of the fatty acid glycerol esters to polyethylene
oxide-
containing fatty acid esters is about 40:60 v/v. In one embodiment, the ratio
of the fatty
acid glycerol esters to polyethylene oxide-containing fatty acid esters is
about 50:50 v/v.
In one embodiment, the ratio of the fatty acid glycerol esters to polyethylene

oxide-containing fatty acid esters is about 60:40 v/v. In one embodiment, the
ratio of the
fatty acid glycerol esters to polyethylene oxide-containing fatty acid esters
is about
70:30 v/v.
In one embodiment, the amphotericin B formulations of the invention include
(a) amphotericin B;
(b) oleic acid glycerol esters; and
(c) lauric acid esters of polyethylene glycol.
In another embodiment, the amphotericin B formulations of the invention
include
(a) amphotericin B;
(b) oleic acid glycerol esters; and
(c) palmitic and stearic acid esters of polyethylene glycol.
In a further embodiment, the amphotericin B formulations of the invention
include
-20-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
(a) amphotericin B;
(b) oleic acid glycerol esters; and
(c) stearic acid esters of polyethylene glycol.
In one embodiment, the amphotericin B formulation of the invention includes
amphotericin B, PECEOL , and GELUCIRE 44/14. In another embodiment, the
amphotericin B formulation of the invention includes amphotericin B, PECEOL ,
and
GELUCIRE 50/13. In a further embodiment, the amphotericin B formulation of
the
invention includes amphotericin B, PECEOL , and GELUCIRE 53/10. In these
embodiments, the ratio of PECEOL to GELUCIRE can be from 20:80 to 80:20
(e.g., 20:80, 30:70; 40:60; 50:50; 60:40; 70:30; and 80:20).
The preparation and characterization of representative amphotericin B
formulations of the invention that include polyethylene oxide-containing fatty
acid esters
is described in Example 1.
The amphotericin B formulations that include polyethylene oxide-containing
fatty
acid esters include amphotericin B that is both partially solubilized
(dissolved) and
present as solid particles to provide a fine solid dispersion. Dispersion of
the
formulations in aqueous media provides a nano-/microemulsion.
The preliminary SEDDS formulations of amphotericin B (see Example 1) did
produce self-emulsification and a small droplet size upon dispersion into
physiological
saline. The dispersion properties of the CAPTEX 355-based formulations were
similar
to those based on mixtures of PECEOL /GELUCIRE 44/14 or 50/13, generating
multiple subpopulations of emulsion droplets in the submicron or 1 pm range
(see
Tables 1 and 2). This particle size would be appropriate for dispersing the
drug in the
GI tract to best facilitate absorption.
The solubility of representative amphotericin B formulations of that include
polyethylene oxide-containing fatty acid esters is illustrated in FIGURES 10A-
10C.
FIGURES 10A, 10B, and 10C compare AmpB concentration (mg/mL) in representative

AmpB formulations of the invention (AmpB/PECEOL GELUCIRE 44/14;
AmpB/PECEOL /GELUCIRE 50/13; and AmpB/PECEOL /GELUCIRE 53/10) at
varying ratios of PECE00:GELUCIRE (60:40; 50:50; and 40:60 v/v) at 2, 4 and
24hrs
(AmpB measured by UV absorbance of centrifuged samples after specified time at
45 C).
FIGURE 11 compares AmpB concentration (% original concentration, 5mg/mL) over
time (0, 1, 5, 7, 15, 21, 28, 36, 43, 49, and 56 days) for an AmpB/PECEOL
formulation
-21-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
(designated PECEOL ) and representative AmpB formulations of the invention
(AmpB/PECEOL /GELUCIRE 44/14, 50/50; and AmpB/PECEOC/DSPE-PEG-2000,
15 mM DSPE-PEG-2000) (AmpB measured by UV absorbance of centrifuged samples
after specified time at 43 C).
Of the formulations evaluated, the
AmpB/PECEOL /GELUORE 44/14, 50/50, formulation shows the greatest stability,
up
to 21 days.
To determine their effectiveness as orally administered formulations, the
stability
of representative amphotericin B formulations of the invention was evaluated
in
simulated gastric fluid.
FIGURE 12 compares AmpB concentration (% original concentration, 5mg/mL)
over time (10, 30, 45, 60, 90, and 120min) in simulated gastric fluid (SGF)
for an
AmpB/PECEOL formulation and representative AmpB formulations of the invention

(AmpB/PECEOL /GELUCIRE 44/14, 50/50; and AmpB/PECEOC/DSPE-PEG-2000,
mM DSPE-PEG-2000) (AmpB measured by UV absorbance of centrifuged samples
15 after specified time at 37 C in SGF, 30mM NaC1 at pH 1.2).
FIGURE 13 compares AmpB concentration (% original concentration, 5mg/mL)
over time (10, 30, 45, 60, 90, 120, and 240min) in fed-state simulated
intestinal fluid
(FeSSIF) for an AmpBIPECEOL formulation and a representative AmpB formulation
of
the invention (AmpB/PECEOL /GELUCIRE 44/14, 50/50) (AmpB measured by UV
absorbance of centrifuged samples after 4hrs in FeSSIF, which contains
potassium
chloride (15.2g/L), sodium taurolaurate (15mM), egg phosphatidylcholine
(3.75mM), and
acetic acid, adjusted to pH 5.0). The representative formulation of the
invention
demonstrates consistent AmpB concentration for up to 2 hours.
FIGURE 14 compares AmpB concentration (% original concentration, 5mg/mL)
over time (10, 30, 45, 60, 90, 120, and 240min) in fed-state simulated
intestinal fluid
(FeSSIF) with enzyme for an AmpB/PECEOL formulation and a representative AmpB

formulation of the invention (AmpB/PECEOC/GELUCIRE 44/14, 50/50) (AmpB
measured by UV absorbance of centrifuged samples after 4hrs in FeSSIF, which
contains
potassium chloride (15.2g/L), sodium taurolaurate (7.5mM), egg
phosphatidylcholine
(2.0mM), glyceryl monooleate (5.0mM), sodium oleate (0.8mM), pancreatin
(1000 u lipase/L), and acetic acid, adjusted to pH 5.8). The representative
formulation of
the invention demonstrates consistent AmpB concentration for up to 2 hours.
-22-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
FIGURE 15 compares AmpB concentration (% original concentration, 5mg/mL)
over time (10, 30, 45, 60, 90, 120, and 240min) in fasted-state simulated
intestinal fluid
(FaSSIF) for an AmpB/PECEOLc) formulation and a representative AmpB
formulation of
the invention (AmpB/PECEOL /GELUCIRE 44/14, 50/50) (AmpB measured by UV
absorbance of centrifuged samples after 4hrs in FaSSIF, which contains
potassium
chloride (7.7g/L), dibasic potassium phosphate (3.9g/L), sodium taurolaurate
(3.0mM),
egg phosphatidylcholine (0.75mM), and acetic acid, adjusted to pH 6.5). The
representative formulation of the invention demonstrates consistent AmpB
concentration
for up to 2 hours.
Each of the evaluated representative formulations of the invention
demonstrated
stability in the simulated fluids over the time period evaluated. The
stability of the
representative amphotericin B formulations in the GI fluids demonstrates their
suitability
as orally administered formulations.
Self-Emulsifying Drug Delivery Systems. The amphotericin B formulations of
the invention can be self-emulsifying drug delivery systems. Self-emulsifying
drug
delivery systems (SEDDS) are isotropic mixtures of oils, surfactants,
solvents, and
co-solvents/surfactants. SEDDS can be used for the design of formulations in
order to
improve the oral absorption of highly lipophilic drug compounds, such as
amphotericin
B. When a SEDDS composition is released into the lumen of the gut, the
composition
disperses to form a fine emulsion, so that the drug remains in solution in the
gut, avoiding
the dissolution step that frequently limits the rate of absorption of
hydrophobic drugs
from the crystalline state. The use of SEDDS usually leads to improved
bioavailability
and/or a more consistent temporal profile of absorption from the gut. A
description of
compositions of SEDDS can be found in C. W. Pouton, Advanced Drug Delivery
Reviews 25: 47-58 (1997).
The amphotericin B formulations of the invention can be orally administered in

soft or hard gelatin capsules and form fine relatively stable oil-in-water
(o/w) emulsions
upon aqueous dilution owing to the gentle agitation of the gastrointestinal
fluids. The
efficiency of oral absorption of the drug compound from the SEDDS depends on
many
formulation-related parameters, such as the formulations' components, polarity
of the
emulsion, droplet size and charge, all of which in essence determine the
self-emulsification ability.
Thus, only very specific pharmaceutical excipient
combinations will lead to efficient self-emulsifying systems.
-23-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
Methods for Administration and Treatment with Amphotericin B. The
administration of intravenous AmpB has been limited by its dose-dependent
kidney
toxicity that has not been predictable by monitoring plasma and/or serum drug
concentration. A number of studies have reported that AmpB, solubilized in
methanol, is
poorly absorbed from the gastrointestinal (GI) tract and therefore is not
commonly
administered orally but intravenously, which can result in the aforementioned
renal
toxicity. However, to date, few studies investigating the development and
assessing the
antifungal activity of oral AmpB formulations have been reported.
The effectiveness of representative amphotericin B formulations of the
invention
that include polyethylene oxide-containing phospholipids in treating fungal
infections is
described in Example 2. The effectiveness of these formulations for treating
Aspergillus
fumigatus and Candida albicans was demonstrated in animal studies.
Treatment of rats infected with Aspergillus fumigatus with representative
amphotericin B formulations of the invention that include polyethylene oxide-
containing
phospholipids significantly decreased total fungal CFU concentrations
recovered in all
the organs added together by 80% compared to non-treated controls (Table 4)
without
significant changes in plasma creatinine levels (Table 5). ABELCET treatment
significantly decreased total fungal CFU concentrations recovered in all the
organs added
together by 88% compared to non-treated controls (Table 4) without significant
changes
in plasma creatinine levels (Table 5).
The results for Candida albicans are similar to those for Aspergillus
fumigatus.
Fungal analysis of the kidneys of Candida albicans-infected rats treated with
a
representative AmpB formulation of the invention demonstrate significantly
decreased
total fungal CFU concentrations compared to control. FIGURE 7 compares Candida
albicans concentration (CFU/ml) in the kidneys of rats infected with Candida
albicans
and treated with control, an AmpB/PECEOL formulation (10mg/kg), a
representative
AmpB formulation of the invention (AmpB/PECEOL /DSPE-PEG-2000, designated
AmpB/DSPE-PEG-2000, 10mg/kg), and intravenous ABELCET (designated AB LC,
5mg/m1). FIGURE 8 compares Candida albicans concentration (CFU/ml) in the
organs
of rats infected with Candida albicans and treated with control, an
AmpB/PECEOL
formulation (10mg/kg), a representative AmpB formulation of the invention
(AmpB/PECEOC/DSPE-PEG-2000, designated AmpB/DSPE-PEG-2000, 10mg/kg),
and intravenous ABELCET (designated ABLC, 5mg/m1). The effectiveness of the
-24-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
representative AmpB formulation in reducing Candida albi cans concentration
was
comparable to ABELCET . Treatment with the representative AmpB formulation
significantly decreased total fungal CFU concentrations recovered in the
kidneys without
significant changes in plasma creatinine levels. FIGURE 9 compares plasma
creatinine
(mg/di) in rats infected with Candida albicans and treated with control, an
AmpB/PECEOL formulation (10mg/kg), a representative AmpB formulation of the
invention (AmpB/PECEOL /DSPE-PEG-2000, designated AmpB/DSPE-PEG-2000,
10mg/kg), and intravenous ABELCET (designated ABLC, 5mg/m1) (blank, Ohr, and
48hr). No renal toxicity was observed as measured by plasma creatine levels.
In another aspect, the invention provides a method for treating an infectious
disease treatable by the administration of amphotericin B.
In the method, a
therapeutically effective amount of an amphotericin B formulation of the
invention is
administered to a subject in need thereof. In one embodiment, the formulation
is
administered orally. In another embodiment, the formulation is administered
topically.
As used herein, the terms "treating" and "treatment" refer to reduction in
severity
and/or frequency of symptoms, elimination of symptoms and/or underlying cause,

reduction in likelihood of the occurrence of symptoms and/or underlying cause,
and
improvement or remediation of damage. Thus, "treating" a patient with an
active agent as
provided herein includes prevention of a particular condition, disease or
disorder in a
susceptible individual as well as treatment of a clinically symptomatic
individual. As
used herein, "effective amount" refers to an amount covering both
therapeutically
effective amounts and prophylactically effective amounts. As used herein,
"therapeutically effective amount" refers to an amount that is effective to
achieve the
desired therapeutic result. A therapeutically effective amount of a given
active agent will
typically vary with respect to factors such as the type and severity of the
disorder or
disease being treated and the age, gender, and weight of the patient.
Infectious diseases treatable by the method and formulations of the invention
include fungal infections (aspergillosis, blastomycosis, candidiasis,
coccidioidomycosis,
crytococcosis, histoplasmosis, mucormycosis, paracoccidioidomycosis, and
sporotrichosis), visceral leishmaniasis, cutaneous leishmaniasis, Chagas
disease, and
Febrile neutropenia. Amphotericin B has been shown to bind to amyloid and
prevent the
formulation of fibrils. Amphotericin B has been indicated as useful for the
treatment of
-25-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
Alzheimer's disease. Accordingly, the amphotericin B formulation of the
invention can
be used in the treatment of Alzheimer's disease.
In summary, in one aspect, the present invention provides amphotericin B
formulations that can be orally administered. The amphotericin B formulations
of the
invention provide excellent drug solubilization, drug stability in simulated
gastric and
intestinal fluids, and have significant antifungal activity without the dose-
limiting renal
toxicity for which the parenteral formulations of amphotericin B are well
known.
Therapeutic Agent Formulations
In another aspect, the present invention provides formulations for the
delivery of
therapeutic agents, methods for making the formulations, and methods for
administering
the therapeutic agents using the formulations.
In one aspect, the invention provides a formulation for the delivery of a
therapeutic agent. The therapeutic agent formulation includes
(a) a therapeutic agent;
(b) one or more fatty acid glycerol esters; and
(c)
one or more polyethoxylated lipids such as one or more polyethylene
oxide-containing phospholipids or one or more polyethylene oxide-containing
fatty acid
esters.
In the therapeutic agent formulation above, the fatty acid glycerol esters,
the
polyethylene oxide-containing phospholipids, and the polyethylene oxide-
containing fatty
acid esters are as described above for the amphotericin B formulations. The
amounts of
these components in the above therapeutic agent formulation is also as
described above
for the amphotericin B formulations. The therapeutic agent can be present in
the
formulation in an amount from about 0.1 mg/mL to about 25 mg/mL of the
formulation.
In certain embodiments, the formulations can further include glycerol in an
amount less
than about 10% by weight.
The therapeutic drug formulation of the invention advantageously solubilizes
difficultly soluble therapeutic drugs. Representative therapeutic agents that
can be
advantageously formulated and delivered by the formulation and methods of the
invention include anticancers, antibiotics, antiviral drugs, antimycotics,
anti-prions,
anti-amoebics, non-steroidal anti-inflammatory drugs, anti-allergics,
immunosuppressive
agents, coronary drugs, analgesics, local anesthetics, anxiolytics, sedatives,
hypnotics,
migraine relieving agents, drugs against motion sickness, and anti-emetics.
-26-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
Specific therapeutic agents that can be advantageously formulated and
delivered
by the formulation and methods of the invention include tetracycline,
doxycycline,
oxytetracycline, chloramphenicol, erythromycin, acyclovir, idoxuridine,
tromantadine,
miconazole, ketoconazole, fluconazole, itraconazole, econazole, griseofulvin,
amphotericin B, nystatine, metronidazole, metronidazole benzoate, tinidazole,
indomethacin, ibuprofen, piroxicam, diclofenac, disodium cromoglycate,
nitroglycerin,
isosorbide dinitrate, verapamile, nifedipine, diltiazem, digoxine, morphine,
cyclosporins,
buprenorphine, lidocaine, diazepam, nitrazepam, flurazepam, estazolam,
flunitrazepam,
triazolam, alprazolam, midazolam, temazepam lormetazepam, brotizolam,
clobazam,
clonazepam, lorazepam, oxazepam, busiprone, sumatriptan, ergotamine
derivatives,
cinnarizine, anti-histamines, ondansetron, tropisetron, granisetrone,
metoclopramide,
disulfiram, vitamin K, paclitaxel, docetaxel, camptothecin, 5N38, cisplatin,
and
carboplatin.
In certain embodiments, the therapeutic agent formulation of the invention can
include a second therapeutic agent.
The therapeutic agent formulation can be a self-emulsifying drug delivery
system.
In one embodiment, the therapeutic agent formulation includes
(a) a therapeutic agent;
(b) one or more fatty acid glycerol esters (e.g., oleic acid glycerol
esters); and
(c) one or more polyethylene oxide-containing phospholipids (e.g., a
distearoylphosphatidyl ethanolamine polyethylene glycol salt).
In one embodiment, the therapeutic agent formulation of the invention includes
a
therapeutic agent, PECEOL , and a distearoylphosphatidyl ethanolamine
polyethylene
glycol salt. In this embodiment, the distearoylphosphatidyl ethanolamine
polyethylene
glycol salt is present in an amount up to about 30 mM.
In another embodiment, the therapeutic agent formulation includes
(a) a therapeutic agent;
(b) one or more fatty acid glycerol esters (e.g., oleic acid glycerol
esters); and
(c) one or more polyethylene oxide-containing fatty acid esters (e.g.,
lauric,
palmitic and/or stearic acid esters of polyethylene glycol).
In one embodiment, the therapeutic agent formulation of the invention includes
a
therapeutic agent, PECEOL , and GELUCIRE 44/14. In another embodiment, the
formulation includes a therapeutic agent, PECEOL , and GELUCIRE 50/13. In a
-27-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
further embodiment, the formulation includes a therapeutic agent, PECEOL , and

GELUCIRE 53/10. In these embodiments, the ratio of PECEOL to GELUCIRE can
be from 20:80 to 80:20 (e.g., 20:80, 30:70; 40:60; 50:50; 60:40; 70:30; and
80:20).
The formulation of two representative therapeutic agents, econazole and
docetaxel, is described in Examples 3 and 4, respectively.
In another aspect, the invention provides a method for administering a
therapeutic
agent. In the method, a therapeutically effective amount of the therapeutic
agent is
administered using the therapeutic agent formulation described above.
In one
embodiment, the formulation is administered orally. In another embodiment, the
formulation is administered topically.
In further aspects, the invention provides methods for treating conditions and

diseases treatable by therapeutic agents formulated in accordance with the
present
invention. In the methods, an effective amount of a therapeutic drug
formulation of the
invention is administered to a subject in need thereof. The methods for
treating
conditions and diseases use formulations of the therapeutic agent families and
specific
therapeutic agents disclosed herein.
Therapeutic Drug Carrier
In a further aspect, the present invention provides compositions for
formulating a
therapeutic agent, methods for making the composition, and methods for
formulating a
therapeutic agent for delivery using the composition.
In one aspect, the invention provides a composition for formulating a
therapeutic
agent for delivery. The composition includes
(a) one or more fatty acid glycerol esters; and
(b) one or more polyethoxylated lipids such as one or more polyethylene
oxide-containing phospholipids or one or more polyethylene oxide-containing
fatty acid
esters.
In the composition above, the fatty acid glycerol esters, the polyethylene
oxide-containing phospholipids, and the polyethylene oxide-containing fatty
acid esters
are as described above for the amphotericin B formulations. The amounts of
these
components in the above composition is also as described above for the
amphotericin B
formulations. In certain embodiments, the compositions can further include
glycerol in
an amount less than about 10% by weight.
-28-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
The composition advantageously solubilizes difficultly soluble therapeutic
drugs
for their delivery. With the incorporation of a therapeutic agent, the
composition can be
provide a self-emulsifying drug delivery system.
In one embodiment, the composition includes
(a) one or more fatty acid glycerol esters (e.g., oleic acid glycerol
esters); and
(b) one or more polyethylene oxide-containing phospholipids (e.g.,
a
distearoylphosphatidyl ethanolamine polyethylene glycol salt).
In one embodiment, the composition includes PECEOL and a
distearoylphosphatidyl ethanolamine polyethylene glycol salt. In this
embodiment, the
distearoylphosphatidyl ethanolamine polyethylene glycol salt is present in an
amount up
to about 30 mM.
In another embodiment, the composition includes
(a) one or more fatty acid glycerol esters (e.g., oleic acid glycerol
esters); and
(b) one or more polyethylene oxide-containing fatty acid esters (e.g.,
lauric,
palmitic and/or stearic acid esters of polyethylene glycol).
In one embodiment, the composition includes PECEOL and GELUCIRE 44/14.
In another embodiment, the composition includes PECEOL and GELUCIRE 50/13.
In
a further embodiment, the composition includes PECEOL and GELUCIRE 53/10. In

these embodiments, the ratio of PECEOL to GELUCIRE can be from 20:80 to
80:20
(e.g., 20:80, 30:70; 40:60; 50:50; 60:40; 70:30; and 80:20).
In another aspect, the invention provides a method for making a therapeutic
agent
formulation. In one embodiment of the method, a therapeutic agent is combined
with the
composition described above. In another embodiment of the method, a
therapeutic agent
is combined with one of the components of the composition (e.g., one or more
fatty acid
glycerol esters) to provide a first combination followed by combining the
first
combination with the other component of the composition (e.g., one or more
polyethylene
oxide-containing phospholipids, or one or more polyethylene oxide-containing
fatty acid
esters).
The formulations and compositions of the invention described herein include
(i.e., comprise) the components recited. In certain embodiments, the
formulations and
compositions of the invention include the recited components and other
additional
components that do not affect the characteristics of the formulations and
compositions
(i.e., the formulations and compositions consist essentially of the recited
components).
-29-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
Additional components that affect the formulations' and compositions'
characteristics
include components such as additional therapeutic agents that
disadvantageously alter or
affect therapeutic profile and efficacy of the formulations or compositions,
additional
components that disadvantageously alter or affect the ability of the
formulations and
compositions to solubilize the recited therapeutic agent components, and
additional
components that disadvantageously alter or affect the ability of the
formulations and
compositions to increase the bioavailability of the recited therapeutic agent
components.
In other embodiments, the formulations and compositions of the invention
include only
(i.e., consist of) the recited components.
The following examples are provide for the purpose of illustrating, not
limiting,
the invention.
EXAMPLES
Materials
The following materials were used as described in the following examples.
Amphotericin B (from Streptomyces sp., Calbiochem, >86% purity) was
purchased from EMD Biosciences (San Diego, CA) and used without further
purification.
Amphotericin B as the commercially available deoxycholate micelle dispersion
(FUNGIZONE ) was purchased from Vancouver General Hospital pharmacy.
Phospholipids and poly(ethylene glycol)-lipids were all from Avanti Polar
Lipids
(Alabaster, AL). HPLC grade solvents were from Fluka. PECEOL (glyceryl
oleate),
LABRASOL (caprylocaproyl macrogol glycerides) and GELUCIRE 44/14,
GELUCIRE 50/13, and GELUCIERE 53/10 were obtained from Gattefosse Canada
(Mississauga, Ontario). CAPTEX 355 and CAPMUL were obtained from Abitech.
Simulated gastric fluid (SGF) without enzymes was composed of 30mM NaC1,
titrated to
pH 1.2 with 1N HC1. Simulated intestinal fluid with pancreatin enzymes (SIFe)
was
prepared according to the US Pharmacopeia method (U5P28) as modified by
Vertzoni et
al., Dissolution media simulating the intralumenal composition of the small
intestine:
physiological issues and practical aspects, J. Pharmacy and Pharm. 56(4):453-
462 (2004),
and was composed of 0.2M NaOH, 6.8g/L of monobasic potassium phosphate and 10
g/L
of pancreatin (Sigma), adjusted to pH 7.5 with NaOH. Fasted-state simulated
intestinal
fluid with bile salts (FaSSIF) (Vertzoni et al.) was composed of 3 mM sodium
taurocholate (Sigma), 3.9g/L sodium dihydrogen phosphate, 6.2g/L NaCl in
water, either
with or without 0.75mM lecithin and then titrated to pH 6.5 with NaOH. Water
was
-30-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
purified by a reverse osmosis system and filtered (0.2pm) prior to use. All
other
chemicals were of reagent grade purchased from SigmaAldrich.
EXAMPLE 1
The Preparation and Characterization of Representative Amphotericin B
Formulations
In this example the preparation and characterization of representative
amphotericin formulations of the invention are described.
Preparation of self-emulsifying drug delivery systems (SEDDS). Amphotericin B
(AmpB) was mixed with the SEDDS lipid vehicles by combining the drug powder
with
the lipids followed by mild heating and stirring (45 C for 1-2h), protected
from light.
Any visible remaining drug particulates were removed by centrifugation at
10,000 xg for
15min.
Preparation of AmpB/PECEOL /DSPE-PEG formulations.
AmpB was
completely dissolved in a mixture of PECEOL at 5mg/mL to which 95% ethanol
(1:3 v/v) had been added, as well as 15 mM distearoyl phosphatidylethanolamine
(DSPE)-poly(ethylene glycol), (PEG) (where n is the average PEG molecular
weight,
350, 550, 750 or 2000). AmpB concentration was 3 mg/ml in the ethanol and 5
mg/mL in
the PECEOL , respectively, to allow for complete drug solubilization in the
initial
mixture. The solution was stirred at 40 C for 1 h, protected from light, to
dissolve AmpB
and lipids, followed by solvent evaporation at 40 C with under vacuum (65
mbar) over
several hours in a rotary evaporator. Ethanol was considered to be completely
removed
by achieving the original weight of the sample containing AmpB, PECEOL , and
lipids
measured immediately prior to the addition of the ethanol. A translucent
yellow mixture
without particulates was formed. No degradation or spectral shape changes of
AmpB
were observed following this processing.
Characterization of amphotericin B stability. Drug concentrations were
measured
by reverse-phase HPLC/UV or by UV spectrophotometry (k=407 nm). For HPLC
analysis of AmpB, samples were diluted in 20% (v/v) methanol in DMSO and 20pL
were
injected on a Luna 5pm (2.0 x 150mm) C18 column (Phenomenex) at 30 C. The
mobile
phase was 10 mM sodium acetate and acetonitrile using a gradient program on a
Waters
996 HPLC system, detected by a Waters photodiode array detector (k=408nm). Run
time
was 13 min and retention time was approximately 8.5 min. Stability of AmpB
against
decomposition or superaggregation upon mild heating of the lipid vehicles
during drug
-31-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
solubilization and upon storage (21 C) over 14 days was assessed by UV
spectral shift
analysis using a Thermoscan UV/visible spectrophotometer with A, = 250-500 nm.

Solubility, physical stability, and self-emulsification. SEDDS (self-
emulsifying
drug delivery systems) formulations had AmpB solubilities ranging from 100-500
g/mL
as measured by HPLC, compared to negligible solubility in aqueous solution (pH
7). For
the tested self-emulsifying lipid mixtures, 3 mg of AmpB powder were combined
with
0.3 ml (10 mg/mL) of the various lipid combination and the mixture was stirred
in a 1 mL
amber glass vial at 37 C for 2h. Following mixing, the samples were
centrifuged at
10,000 x g for 15 min to remove any remaining drug particulates. This
procedure does
not sediment the lipid components. The samples were then dispersed as a 1:1000
(v/v)
dilution in 150mM NaC1 with vigorous mixing at 37 C for 30 min.
The results for CAPTEX 355-based AmpB SEDDS are summarized in Table 1.
The results for PECEOL /GELUC1RE -based AmpB SEDDS are summarized in
Table 2.
Table 1. CAPTEX 355-based Amphotericin B Preliminary SEDDS.
Effective Hydro- Subpopulations
Components (% v/v) dynamic Relative
Diameter (nm) Diameter range pro-
portion
0 0
CAPTEX Tween CAPMUL NaH2PO4 (Poly-dispersity (nm)
355 80 MCM (10mM, pH index)
4.0)
50 17 31 2 186 (0.232) 49-69 79
196-277 21
53 5 40 2 237 (0.278) 24-44 73
111-242 12
614-1334 15
58 10 30 2 216 (0.258) 44-64 51
154-255 5
541-893 44
63 5 30 2 223 (284) 62-89 19
168-241 6
413-648 75
70 10 18 2 168 (0.215) 50-65 82
199-273 18
50 17 31 2 179 (0.24) 55-71 80
214-297 20
-32-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
Particle sizing was performed after mixture was dispersed 1:1000 (v/v) in
150 mM NaC1 at 37 C x 30 min. Relative proportion is based on the cumulative
distribution of particle sizes.
Table 2. PECEOL /GELUCIRE -based Amphotericin B SEDDS.
Components (% v/v) Effective Hydrodynamic Sub-populations Relative
PECEOL GELUCIRE Diameter (nm) (nm) proportion
44/14 (polydispersity)
70 30 156 20-43 55
(0.291) 75-132 16
307-621 29
50 50 314 35-62 56
(0.319) 165-332 5
768-1345 59
30 70 252 28-50 63
(0.294) 135-241 7
568-1160 30
PECEOL GELUCIRE
50/13
70 30 158 30-47 55
(0.279) 94-168 17
336-599 28
50 50 192 19-22 25
(0.301) 71-145 24
396-704 51
30 70 396 50-99 25
(0.265) 228-527 63
1703-2382 12
Particle sizing was performed after mixture was dispersed 1:1000 (v/v) in
150mM
NaC1 at 37 C x 30min. Relative proportion is based on the cumulative
distribution of
particle sizes.
As shown in Tables 1 and 2, the effective hydrodynamic diameter was
168-237 nm at equilibrium for CAPTEX 355/CAPMUL MCM/Tween 80 (Table 1)
and 58-396 nm for PECEOL /GELUCIRE 44/14 (Table 2) but not for formulations
based on soybean oil, PECEOL /LABRASOL , or PECEOL /GELUCIRE 50/13
(>1 mm, data not shown). Importantly, multiple subpopulations of emulsion
droplet size
were observed. For the CAPTEX 355-based SEDDS, these subpopulations included
-33-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
diameters consistent with the size of micelles (e.g. 20-50nm) and all
populations
remained in the submicron range (Table 1). For the PECEOL /GELUCIRE 44/14
mixtures, three subpopulations were observed, including 20-50nm, 1000-200nm
range
(minor population) and those closer to 1 pm in diameter. The proportion of
very small
and large droplets varied by ratio of components. In the case of PECEOL
/GELUCIRE
50/13, three subpopulations were also observed with similar droplet size
ranges as with
PECEOC/GELUCIRE 44/14, although there was a trend to slightly larger droplets
in
the largest diameter subpopulation, which also increased with increasing
proportion of
GELUCIRE 50/13 (Table 2). Representative single samples are fully described
in the
tables, however, it should be noted that replicate samples did show
consistency in their
effective diameters and in the particle size ranges of the subpopulations.
Visual
observations were made regarding miscibility, phase separation and
precipitation over
several days at ambient temperature (21 C). Several SEDDS formulations
remained
transparent and homogeneous. For example, the CAPTEX -based SEDDS formulations
all generated semi-transparent mixtures after mixing with 150 mM NaC1 that
were
homogeneous for all combination ratios of CAPTEX , Tween 80 and sodium
phosphate
(Table 1). Combinations of PECEOL and GELUCIRE 44/14 in the range of 70/30
to
30/70 (v/v) generated a fine emulsion whereas some partial solidification was
observed
over 24h at 21 C when using PECEOL with GELUCIRE 50/13, consistent with the
high melting point of GELUCIRE 50/13 (50 C).
AmpB Solubility in PECEOL /DSPE-PEG Formulations and Emulsification in
Fasted-State Simulated Intestinal Fluid. The combination of PECEOL and DSPE-
PEG,
where average molecular weight of PEG was varied from 350 to 2000, showed an
even
greater solubilization of AmpB (5 mg/mL) compared to the preliminary SEDDS
formulations. At concentrations >10mg/mL, some precipitation of AmpB did occur
upon
standing at ambient temperature (21 C) over 24h. Upon dispersion in SGF at 37
C at
0.5 mg/mL followed by stirring for 30min, the PECEOL /DSPE-PEG2000 AmpB
formulations generated translucent emulsions with particle sizes of 300-500 nm
and no
visible precipitate. In some cases, there appeared to be two populations of
submicron
particles, with some at 100 nm and others several hundred nm in diameter.
AmpB Stability in Simulated Gastric and Intestinal Fluids. Amphotericin B in
PECEOC/DSPE-PEG formulations (5 mg/mL) were prepared in triplicate and were
incubated in simulated gastric fluid (SGF) as a 1:10 (v/v) dilution) or in
simulated
-34-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
intestinal fluid prepared with and without lecithin or with enzymes (as
described above)
as a 1:50 (v/v) dilution at 37 C with vigorous stirring. Incubation times were
0, 10, 30 or
120 mm. At each time point, AmpB concentration was determined by
spectrophotometry
using triplicate measures of absorbance (407 nm) after complete solubilization
in 95%
ethanol to clarify the samples, thereby also diluting the samples to the
linear range of the
UV assay. Values were normalized to the baseline at 330nm and concentrations
were
calculated based on an amphotericin B standard curve prepared in each fluid
type
(r2>0.99). The linearity of the standard curve and concentration range of
standards
prepared in PECEOL /DSPE-PEG were not affected by the type of simulated GI
fluid or
by incubation time, however, separate triplicate standard curves were prepared
for each
formulation containing the various molecular weights of DSPE-PEG (350, 550,
750, or
2000).
The chemical stability and aggregation state (monomeric vs. self-associated)
of
AmpB was evaluated in USP simulated gastric fluid as well as fasted-state
simulated
intestinal fluid with and without bile salts and pancreatin. As described
above, AmpB in
PECEOL alone or in PECEOC/DSPE-PEG formulations (PEG molecular
weight = 350, 550, 750, or 2000) was prepared at 5mg/mL and incubated in the
simulated
GI fluids for a total period of 2h. At 30 mm intervals, the AmpB concentration
and UV
spectra were evaluated. AmpB exhibits 5 main spectrophotometric peaks in the
UV
range. Peaks 4 and 5 have the greatest amplitude in monomeric AmpB, whereas
there is
a left shift when AmpB becomes self-associated. FIGURE 3A shows typical UV
spectra
of AmpB in PECEOL /DSPE-PEG over the linear range of the UV assay,
illustrating the
predominance of monomeric AmpB. This pattern was maintained when PEG molecular

weight was varied from 350 to 2000 (data not shown). The same spectral pattern
was
also observed following incubation in SGF, as well as resulting in nearly
identical
standard curves for the various AmpB/PECEOL /DSPE-PEGn preparations, as shown
in
FIGURE 3B.
Regarding chemical stability, the trend was to slightly less drug stability in

formulations prepared with DSPE-PEG 350 or 550 compared to DSPE-PEG 750 or
2000.
AmpB alone (e.g., neat powder) was not soluble in these media and therefore
could not
be used properly as a control at comparable concentrations due to the
confounding factors
of increased dissolution over time vs. degradation. AmpB in PECEOL alone
prepared
otherwise the same way was included as a negative control for the stabilizing
effect of
-35-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
DSPE-PEG in the formulations. AmpB/PECEOL showed a trend to slightly less
drug
stability in SGF than formulations containing DSPE-PEG 350 or 2000 as shown in

FIGURE 4. FIGURE 5 shows AmpB stability in simulated intestinal fluid
containing bile
salts either without lecithin (FIGURE 5A) or with lecithin (FIGURE 5B) is less
for
AmpB in PECEOL alone or in PECEOL /DSPE-PEG 350 compared to formulations
using the higher PEG molecular weights.
FIGURE 6 illustrates the stability of AmpB in simulated intestinal fluid with
pancreatin, which contains degradative enzymes. These data suggest better
stability of
formulations containing DSPE-PEG 750 or 2000 compared to 350 or 550 or in
PECEOL
alone. In evaluating the degradation of AmpB in PECEOL alone, however, is it
is
important to note that poor mixing of AmpB/PECEOL in the simulated GI fluids
was
observed; this formulation tended to float. No changes associated with
conversion of the
monomeric form vs. aggregated AmpB, such as a difference in the height ratios
of
specific peaks in the UV spectra or overall pattern, was observed following
the full
incubation time in the various media described here (data not shown).
Particle size analysis.
Particle size analysis by dynamic light scattering
(ZetaPALS instrument, Brookhaven Laboratories, New York, operating at 650nm)
was
used to assess self-emulsification properties. Emulsion droplet size was
measured in
physiological saline (150mM NaC1) following 30min incubation at 37 C.
For
PECEOC/DSPE-PEG AmpB formulations, the mean diameter was measured at 37 C
every 10min in preliminary experiments and it was found that the mean diameter
came to
equilibrium by lh and remained stable. Drug stability was measured after 2h,
therefore
2h was the time point used for reported particle size analysis from samples
incubated in
simulated intestinal fluids. Two data analysis modes are available in the
ZetaPALS
software (version 3.88), which calculate a weighted mean effective
hydrodynamic
diameter based on a lognormal distribution, and a multimodal distribution to
identify
subpopulations centered on two or more mean diameters. Both values are
reported where
bimodal or multimodal distributions were detected, with the proportion of each

subpopulation reported based on the cumulative distribution analysis (ZetaPALS
software, version 3.88).
Varying the DSPE-PEG molecular weight had no clear effect on the emulsion
droplet size in simulated intestinal fluid (Table 3) following mixing over a
period of 2h at
37 C. Submicron mean diameters were observed in the range of 300-600nm with a
fairly
-36-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
wide polydispersity. A bimodal particle size distribution was also generated,
with a small
subpopulation centered in submicron range (150-300nm) and another centered in
the
1-2pm range. AmpB in PECEOL alone also formed droplets of similar size and
distribution in simulated intestinal fluid. These particle size measurements
were
performed in the absence of lecithin, which formed very large emulsion
droplets under
the mixing conditions employed and opacified the samples. The results are
presented in
Table 3.
Table 3. PECEOC/DSPE-PEG Amphotericin B Formulation Particle Size.
Formulation: Effective Diameter Polydispersity index Sub-populations
Relative
PECEOL /DSPE-PEGn (nm) (nm)
proportion
n lognormal
distribution
350 370 0.344 129-186 20
888-1282 80
550 600 0.402 108-171 20
1206-1909 80
750 596 0.395 134-210 18
1245-3400 82
2000 533 0.392 119-200 30
1390-2330 70
AmpB in PECEOL 351 0.333 128-194 20
alone 738-1120 80
Particle sizing by dynamic light scattering of AmpB in PECEOL /DSPE-PEG,
where the molecular weight of PEG was varied from 350 to 2000, following 2h
incubation simulated fasted-state intestinal fluid (pH 6.8) at 0.5mg AmpB/mL.
Relative
proportion is based on the cumulative distribution of particle sizes.
EXAMPLE 2
The Effectiveness of Representative Amphotericin B Formulations
in Treating Fungal Infections: Aspergillus fumigatus and Candida albicans
In this example, the effectiveness of representative amphotericin B
formulations
of the invention in treating fungal infections is described. Animal studies
were conducted
to determine the effectiveness of representative amphotericin B formulations
of the
invention in treating rats infected with Aspergillus fumigatus or Candida
albicans.
1.
Aspergillus fumigatus (2.7-3.3 x 107 colony forming units [CHID was
injected via the jugular vein; 48h later male albino Sprague-Dawley rats (350-
400 g) were
-37-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
administered either as a single oral gavage of monoglyceride-DSPE/PEG2000-
based
AmpB (10 mg AmpB/kg; n=7) twice daily for 2 consecutive days, a single
intravenous
(i.v.) dose of ABELCET (5 mg AmpB/kg; n=4), or physiologic saline (non-
treated
controls; n=9) once daily for 2 consecutive days. Organs were harvested at
sacrifice
(day 3) and processed (see below). Blood was drawn before inoculation (Blank),
pre-dose (0 hour) and 48 hours after treatment for plasma creatinine analysis.
Male
albino Sprague-Dawley rats (350 to 400g) were purchased from Charles River
Laboratories (Wilmington, MA). The rats were surgically implanted with a port
(Access
Technologies) and catheter with access to venous blood by a similar method
used for
rabbits. The rats were housed in an animal care facility with a 12 hour light-
dark cycle
and controlled temperature and humidity. The rats were given ad libitum access
water
and standard rat chow (Purina Rat Chow) for the duration of the study. The
ports were
primed daily with normal saline and heparin to prevent blockages. The animals
were
cared for according to principals promulgated by the Canadian Council on
Animal Care
and the University of British Columbia.
Aspergillus fumigatus Inoculum. A. fumigatus were collected from a pool of
patients with either disseminated aspergillosis (BC Centre for Disease
Control). Cultures
were grown on Sabouraud dextrose agar for 48 hours at 37 C. Conidia were
isolated by
washing the agar with pyrogen free saline. The conidia were suspended by
vortexing
with glass beads and diluted with pyrogen free saline to obtain between 2.7 to
3.3x107conidia in 300 1 of saline. Conidia were counted using a hemocytometer
and a
100 1 aliquot was serially diluted and aliquots were plated on sabouraud
dextrose agar for
48 hours at 37 C to determine the number of viable conidia and purity of the
inoculum.
The average percentage of viable conidia in the inoculum was 62% 19. None of
the
spore suspensions were contaminated with any other organism. Rats were
inoculated
with 300 1 through the indwelling port 48 hours before the beginning of
treatment to
allow for aspergillosis to develop.
2. Candida albicans (1-1.35 x 106 colony forming units [CFU]) was
injected
via the jugular vein; 48h later male albino Sprague-Dawley rats (350-400 g)
were
administered either as a single oral gavage of monoglyceride-DSPE/PEG2000-
based
AmpB (10 mg AmpB/kg; n=7) twice daily for 2 consecutive days, a single
intravenous
(i.v.) dose of ABELCET (5 mg AmpB/kg; n=3), or physiologic saline (non-
treated
controls; n=9) once daily for 2 consecutive days. Organs were harvested at
sacrifice
-38-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
(day 3) and processed (see below). Blood was drawn before inoculation (Blank),

pre-dose (0 hour) and 48 hours after treatment for plasma creatinine analysis.
Male
albino Sprague-Dawley rats (350 to 400g) were purchased from Charles River
Laboratories (Wilmington, MA). The rats were surgically implanted with a port
(Access
Technologies) and catheter with access to venous blood by a similar method
used for
rabbits. The rats were housed in an animal care facility with a 12 hour light-
dark cycle
and controlled temperature and humidity. The rats were given ad libitum access
water
and standard rat chow (Purina Rat Chow) for the duration of the study. The
ports were
primed daily with normal saline and heparin to prevent blockages. The animals
were
cared for according to principals promulgated by the Canadian Council on
Animal Care
and the University of British Columbia.
Candida albicans Inoculum. Candida albicans were collected from a pool of
patients with either disseminated Candidiasis (BC Centre for Disease Control).
Cultures
were grown on Sabouraud dextrose agar for 48 hours at 37 C. Conidia were
isolated by
washing the agar with pyrogen free saline. The conidia were suspended by
vortexing
with glass beads and diluted with pyrogen free saline to obtain between 2.7 to

3.3x107 conidia in 300[0 of saline. Conidia were counted using a hemocytometer
and a
100111 aliquot was serially diluted and aliquots were plated on sabouraud
dextrose agar for
48 hours at 37 C to determine the number of viable conidia and purity of the
inoculum.
The average percentage of viable conidia in the inoculum was 62% 19. None of
the
spore suspensions were contaminated with any other organism. Rats were
inoculated
with 300 1 through the indwelling port 48 hours before the beginning of
treatment to
allow for aspergillosis to develop.
3.
Animal Methods. One-ml whole blood samples were drawn into pediatric
collection tubes (3.6mg K2 EDTA) before infection (blank), pre-dose (0 hour)
and
48 hours after treatment (48 hour). All whole blood samples were mixed by
inversion
and plasma was separated by centrifugation (15 minutes, 3000 RPM at 4 C).
Plasma
samples were stored at -20 C for creatinine analysis. After the collection of
the 48 hour
blood specimen, the rat was euthanized with intravenous overdose (1 me of
EUTHANYL , (sodium pentobarbital 240 mg/ml). Spleen, right kidney, liver,
lung,
heart and brain tissue samples were harvested, weighed and placed in sterile
containers.
Normal saline was added, 1 ml/g of specimen and homogenized (Heidolph diax
900). An
-39-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
aliquot of organ homogenate was stored at room temperature until plating and
the
remaining sample was placed at -80 C until HPLC analysis.
The choice of organ colony forming units (CFU) as an indicator of antifungal
activity was based on previously published work (K.M. Wasan et al., Assessing
the
antifungal activity and toxicity profile of Amphotericin B Lipid Complex
(ABLC;
ABELCET ) in combination with Caspofungin in experimental systemic
aspergillosis,
Journal of Pharrn. Sci. 2004; 93(6):1382-1389). Aliquots of 100 I full
strength organ
homogenate and 1:10 dilution (with sterile saline) were each spread plated
onto Saboraud
Dextrose Agar plates in duplicate. After 48 hr incubation at 37 C, the
resulting colonies
of A. fumigatus or C. albicans were counted and averaged over the duplicate
plates. The
limit of detection of the assay was 0.1 x 102 CFU/ml homogenate.
Renal toxicity was indirectly assessed, as previously described (K.M. Wasan
et al., Assessing the antifungal activity and toxicity profile of Amphotericin
B Lipid
Complex (ABLC; ABELCET ) in combination with Caspofungin in experimental
systemic aspergillosis, Journal of Pharm. Sci. 2004; 93(6):1382-1389), by
determining
creatinine concentration in plasma using a commercially available kit (Sigma
Chemicals
Co.). A baseline was determined by measuring creatinine concentration in the
blank
sample, and was compared to plasma creatinine concentration in the 0 hour (pre-
dose),
48 hour samples. For the purposes of this study, a 50% or greater increase in
plasma
creatinine concentration as compared to baseline was considered to be a sign
of renal
toxicity.
4. Statistical Analysis. The number of CFU's in organs and plasma
creatinine concentrations prior to and following administration of treatment
were
compared between each treatment group by analysis of variance (INSTAT2;
GraphPad
Inc.). Critical differences were assessed by Tukey post hoc tests. Serum
creatinine
values were compared prior to 48 hours following treatment using repeat
measures
ANOVA with a Tukey post hoc test to determine critical differences (Prism 4;
Graphpad
Inc.). A difference was considered significant if the probability of chance
explaining the
results was reduced to less than 5% (p<0.05). All data were expressed as a
mean
standard error of the mean.
Antifungal activity and renal toxicity in rats infected with Aspergillus
fumigatus.
PECEOLciDSPE/PEG2000-based oral AmpB treatment significantly decreased total
fungal CFU concentrations recovered in all the organs added together by 80%
compared
-40-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
to non-treated controls (Table 4) without significant changes in plasma
creatinine levels
(Table 5). ABELCET treatment significantly decreased total fungal CFU
concentrations
recovered in all the organs added together by 88% compared to non-treated
controls
(Table 4) without significant changes in plasma creatinine levels (Table 5).
Table 4. Fungal analysis of Aspergillus fumigatus-infected male Sprague Dawley
rats
treated with oral gavage doses of Normal Saline (non-treated control),
Amphotericin-
DSPE-PEG200 incorporated into PECEOL (10 mg/kg twice daily x 2 days) or a
single
intravenous dose of ABELCET (ABLC; 5 mg/kg once daily x 2 days). All rats
were
infected with 2.9-3.45 x 107 Viable Colony Forming Units (CFU)/0.3 ml/rat of
Aspergillus fumigatus prior to initiation of treatment.
Treatment Infected Tissues (CFU/ml of homogenized tissues)
Groups
Brain Lungs Heart Liver Spleen Kidney All Organs
Non- 3538 74 101 308 1163 364 5549
treated 1810 30 63 114 772 119 2498
Controls
(n=9)
ABLC 5 550 10 15 18 88 10 690
(n=4) 445a 4a 3a 5a 44a Oa 419a
AmpB- 736 51 20 180 107 44 1139
DSPE- 186a 18 4 48 32a 10a 221a
PEG-
2000
(n=7)
ap<0.05 vs. non-treated controls using student T-Test; All Data are presented
as
mean SEM.
*Note: Previous studies have shown that AmpB alone does not have measurable
accumulation at the doses used herein. ABLC: Amphotericin B Lipid Complex.
-41-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
Table 5. Plasma creatinine concentrations before infection (blank), pre-dose
(0 hour) and 48 hours after treatment (48 hour).
Blank 0 48
Creatinine (mg/di) 0.4 0.5 0.9
Control (n=9) 0.1 0.1 0.2
AmpB/DSPE-PEG2000/PECEOL 0.6 0.6 0.5
10mg/kg (oral) (n=7) 0.2 0.2 0.1
0.3 0.4 0.5
ABLC 5mg/kg-IV (n=4) 0.2 0.1 0.1
Data presented as mean SEM
The results for Candida albicans are similar to those for Aspergillus
fumigatus.
Fungal analysis of the kidneys of Candida albicans-infected rats treated with
a
representative AmpB formulation of the invention demonstrate significantly
decreased
total fungal CFU concentrations compared to control. FIGURE 7 compares Candida
albicans concentration (CFU/ml) in the kidneys of rats infected with Candida
albicans
and treated with control, an AmpB/PECEOL formulation (10mg/kg), a
representative
AmpB formulation of the invention (AmpB/PECEOL /DSPE-PEG-2000, designated
AmpB/DSPE-PEG-2000, 10mg/kg), and intravenous ABELCET (designated ABLC,
5mg/m1). FIGURE 8 compares Candida albicans concentration (CFU/ml) in the
organs
of rats infected with Candida albicans and treated with control, an
AmpB/PECEOL
formulation (10mg/kg), a representative AmpB formulation of the invention
(AmpB/PECEOL /DSPE-PEG-2000, designated AmpB/DSPE-PEG-2000, 10mg/kg),
and intravenous ABELCET (designated ABLC, 5mg/m1). The effectiveness of the
representative AmpB formulation in reducing Candida albicans concentration was
comparable to ABELCET . Treatment with the representative AmpB formulation
significantly decreased total fungal CFU concentrations recovered in the
kidneys without
significant changes in plasma creatinine levels.
FIGURE 9 compares plasma creatinine (mg/de in rats infected with Candida
albicans and treated with control, an AmpB/PECEOL formulation (10mg/kg), a
representative AmpB formulation of the invention (AmpB/PECEOL /DSPE-PEG-2000,
designated AmpB/DSPE-PEG-2000, 10mg/kg), and intravenous ABELCET (designated
ABLC, 5mg/m1) (blank, Ohr, and 48hr). No renal toxicity was observed as
measured by
plasma creatine levels.
-42-

CA 02688036 2009-11-24
WO 2008/144888
PCT/CA2008/000975
EXAMPLE 3
Representative Econazole Formulation
In this example, the preparation and characterization of a representative
econazole
formulation of the invention is described. The solubility of econazole in
water is
< lmg/mL (19-66 F), and the solubility in ethanol is < 20mg/mL.
Econazole nitrate formulations (10 and 15 mg econazole nitrate/mL formulation)

were prepared by a method similar to the method for preparing the amphotericin
B
formulation described in Example 1. To econazole nitrate (Sigma) and DSPE-
PEG2000
(15mM) powders was added 45 C PECEOL and the resulting mixture shaken for 2-
4h at
45 C in a shaking incubator. No ethanol was used. Samples were centrifuged in
order to
visualize unsolubilized material. The mixture was then evaluated for clarity
over time.
The product econazole nitrate formulation was incubated at 37 C for 2h in
simulated gastric fluid (1:100 v/v dilution), fasted-state simulated
intestinal fluid, and
fed-state simulated intestinal fluid with and without pancreatin (all at 1:500
v/v dilution)
to evaluate emulsification properties. Emulsion droplet size was evaluated
immediately
by dynamic light scattering (Zetasizer, Malvern Instruments).
The formulations were evaluated for clarity over time. The results are
summarized in Table 6.
Table 6. Visual appearance of econazole formulations.
Econazole Visual Appearance
Formulation
Day 0 24h 4 days 5
days
10mg/mL Clear Clear Clear Clear >5days
15mg/mL Some solids Still clear Not tested Not tested
remaining after portion with
centrifugation solids
Emulsion droplet size analysis was determined for the econazole formulation
(10mg/mL) in simulated gastric and intestinal fluids (SGF, FaSSIF, FeSSIF, and

FeSSIFe), with mean based on peak analysis by volume. Emulsion droplet size in
the
table refers to the mean and peak half-width, a range for each subpopulation.
The results
are summarized in Table 8.
-43-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
Table 7. Emulsion droplet size for econazole formulation.
Media Dilution factor Lower size Upper size Mean (nm)
(nm) (nm)
SGF 100 30 291 72 46 (71%) and
310 200 (29%)
FaSSIF 500 59 333 123 65 (44%) and
342 138 (56%)
FeSSIF 500 66 1557 386 182
(10%);
726 143 (5%) and
2503 565 (85%)
FeSSIF'e 500 510 >5000 650 150(75%)
>5 m (25%)
SGF: simulated gastric fluid
FaSSTF: fasted-state simulated intestinal fluid
FeSSLF: fed-state simulated intestinal fluid
FeSSIFe: fed-state simulated intestinal fluid with pancreatic enzymes (Sigma)
Composition of simulated gastric/intestinal fluids (1L) used in emulsification

studies for the econazole formulation.
SGF (simulated gastric fluid):
Distilled water: 1L
Sodium chloride: 30mM (1740mg/L)
Hydrochloric acid: as required to adjust to pH 1.2.
FaSSIF (fasted-state simulated intestinal fluid):
Dibasic potassium phosphate: 3.9g
Distilled water: 1L
Sodium taurocholate: 3mM (1613.04mg/L)
Egg phosphatidylcholine: 0.75mM (570.07mg/L)
Potassium chloride: 7.7g
Hydrochloric acid: as required to adjust pH to 6.5.
FeSSIF (fed-state simulated intestinal fluid):
Distilled water: 1L
Acetic acid: 8.65g = 9.073m1
Sodium taurocholate: 15mM (8065.2mg/L)
Egg phosphatidylcholine: 3.75mM (2850.34mg/L)
-44-

CA 02688036 2009-11-24
WO 2008/144888 PCT/CA2008/000975
Potassium chloride: 15.2g
Hydrochloric acid or sodium hydroxide: as required to adjust pH to 5Ø
FeSSIF with pancreatic enzymes:
Distilled water: 1L
Sodium taurocholate: 7.5mM (4032.6mg/L)
Egg phosphatidylcholine: 2.0mM (1520.18mg/L)
Glyceryl monooleate: 5.0mM (1782.72mg/L)
Sodium oleate: 0.8mM (241.96mg/L)
Pancreatin: 1000 u lipase
Acetic acid: 9.073m1
Potassium chloride: 15.2g
Hydrochloric acid or sodium hydroxide: as required to adjust pH to 5.8.
EXAMPLE 4
Representative Docetaxel Formulation
In this example, the preparation of a representative docetaxel formulation of
the
invention is described. The solubility of docetaxel in water is about 10-25
lag/mL.
A docetaxel formulation (10mg docetaxel/mL formulation) was prepared by
combining docetaxel (Fluka) powder with the DSPE-PEG2000 powder and wetting
the
combined powders 100% ethanol to 10% v/v of the final intended volume. The
ethanol
did not solubilize the powders. To the wetted powders was added PECEOL that
was
pre-warmed to 50 C, followed by vortex mixing for 2 min which resulted in a
clear
solution. Ethanol was not removed.
Ethanol was used with docetaxel as a co-solvent in this formulation to
maximize
solubility. Docetaxel is poorly water soluble but has polar regions.
The formulation was evaluated for clarity over time. The results are
summarized
in Table 8.
-45-

CA 02688036 2015-05-20
WO 2008/144888 PCT/CA2008/000975
Table 8. Visual appearance of docetaxel formulations.
Docetaxel Visual Appearance
Formulation
Day 0 24h 4 days 5 days
10mg/mL Solidified at 4 C Solidified at 4 C
Solidified at 4 C
at 4 C but clear upon but clear
upon but clear upon
melting briefly melting briefly melting briefly
10mg/mL Clear Clear Clear Clear
at 21 C
10mg/mL Solidified at Solidified at Solidified at
at -20 C -20 C but clear -20 C but clear -20
C but clear
upon melting upon melting upon melting
briefly briefly briefly
10mg/m1 Clear Clear with yellow
Clear with yellow
at 50 C tinged* tinge*
*Consistent with color change observed with PECEOL alone at 50 C for this
length of time.
Docetaxel formulations of the invention include those as described above, but
that
do not include ethanol.
The scope of the claims should not be limited by the preferred embodiment
and examples, but should be given the broadest interpretation consistent with
the
ID description as a whole.
-46-

Representative Drawing
A single figure which represents the drawing illustrating the invention.
Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date 2016-08-16
(86) PCT Filing Date 2008-05-23
(87) PCT Publication Date 2008-12-04
(85) National Entry 2009-11-24
Examination Requested 2013-05-22
(45) Issued 2016-08-16

Abandonment History

There is no abandonment history.

Maintenance Fee

Last Payment of $624.00 was received on 2024-04-09


 Upcoming maintenance fee amounts

Description Date Amount
Next Payment if standard fee 2025-05-23 $624.00
Next Payment if small entity fee 2025-05-23 $253.00

Note : If the full payment has not been received on or before the date indicated, a further fee may be required which may be one of the following

  • the reinstatement fee;
  • the late payment fee; or
  • additional fee to reverse deemed expiry.

Patent fees are adjusted on the 1st of January every year. The amounts above are the current amounts if received by December 31 of the current year.
Please refer to the CIPO Patent Fees web page to see all current fee amounts.

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $400.00 2009-11-24
Maintenance Fee - Application - New Act 2 2010-05-25 $100.00 2009-11-24
Registration of a document - section 124 $100.00 2010-02-08
Maintenance Fee - Application - New Act 3 2011-05-24 $100.00 2011-05-19
Maintenance Fee - Application - New Act 4 2012-05-23 $100.00 2012-03-01
Maintenance Fee - Application - New Act 5 2013-05-23 $200.00 2013-05-07
Request for Examination $200.00 2013-05-22
Maintenance Fee - Application - New Act 6 2014-05-23 $200.00 2014-05-06
Maintenance Fee - Application - New Act 7 2015-05-25 $200.00 2015-04-22
Maintenance Fee - Application - New Act 8 2016-05-24 $200.00 2016-04-22
Final Fee $300.00 2016-06-03
Maintenance Fee - Patent - New Act 9 2017-05-23 $200.00 2017-05-03
Maintenance Fee - Patent - New Act 10 2018-05-23 $250.00 2018-05-02
Maintenance Fee - Patent - New Act 11 2019-05-23 $250.00 2019-05-01
Maintenance Fee - Patent - New Act 12 2020-05-25 $250.00 2020-04-29
Maintenance Fee - Patent - New Act 13 2021-05-25 $255.00 2021-04-28
Maintenance Fee - Patent - New Act 14 2022-05-24 $254.49 2022-04-27
Maintenance Fee - Patent - New Act 15 2023-05-23 $473.65 2023-04-19
Maintenance Fee - Patent - New Act 16 2024-05-23 $624.00 2024-04-09
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
THE UNIVERSITY OF BRITISH COLUMBIA
Past Owners on Record
WASAN, ELLEN K.
WASAN, KISHOR M.
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Abstract 2009-11-24 2 77
Claims 2009-11-24 8 323
Drawings 2009-11-24 20 983
Description 2009-11-24 46 2,632
Representative Drawing 2009-11-24 1 26
Cover Page 2010-01-27 1 57
Cover Page 2016-06-21 1 57
Claims 2013-05-22 3 84
Claims 2015-01-19 2 53
Claims 2015-05-20 2 54
Description 2015-05-20 46 2,621
Representative Drawing 2016-07-20 1 25
Claims 2015-08-20 2 54
Claims 2015-12-10 2 54
Assignment 2010-02-08 5 156
Correspondence 2010-02-08 3 80
PCT 2009-11-24 3 126
Assignment 2009-11-24 4 110
Correspondence 2010-01-19 1 20
Correspondence 2010-03-24 1 16
Prosecution-Amendment 2013-05-22 1 45
Prosecution-Amendment 2015-05-20 7 245
Prosecution-Amendment 2013-05-22 5 128
Prosecution-Amendment 2013-06-06 1 20
Prosecution-Amendment 2013-06-25 1 32
Prosecution-Amendment 2013-06-24 1 35
Correspondence 2013-07-09 1 13
Prosecution-Amendment 2015-01-19 4 123
Prosecution-Amendment 2014-08-01 2 73
Prosecution-Amendment 2015-03-25 3 221
Examiner Requisition 2015-08-12 3 201
Amendment 2015-08-20 4 133
Examiner Requisition 2015-11-27 3 211
Amendment 2015-12-10 3 104
Final Fee 2016-06-03 1 48